US9068230B2 - Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies - Google Patents

Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies Download PDF

Info

Publication number
US9068230B2
US9068230B2 US12/741,814 US74181408A US9068230B2 US 9068230 B2 US9068230 B2 US 9068230B2 US 74181408 A US74181408 A US 74181408A US 9068230 B2 US9068230 B2 US 9068230B2
Authority
US
United States
Prior art keywords
antibody
lymphoma
sample
expression
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US12/741,814
Other versions
US20110104671A1 (en
Inventor
David Dornan
Bart Burington
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US12/741,814 priority Critical patent/US9068230B2/en
Assigned to GENENTECH, INC. reassignment GENENTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BURINGTON, BART, DORNAN, DAVID
Publication of US20110104671A1 publication Critical patent/US20110104671A1/en
Application granted granted Critical
Publication of US9068230B2 publication Critical patent/US9068230B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates generally to the fields of predicting, assessing, aiding assessment of responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies.
  • CD40 is a type I transmembrane protein of the tumor necrosis receptor superfamily. CD40 is an important molecule involved in B-cell proliferation and differentiation, immunoglobulin isotype switching, and cell viability. Receptor signaling is initiated by the binding of CD40 to the CD40 ligand (CD40L or CD154), which is primarily expressed on activated CD4+ T cells.
  • CD40L or CD154 CD40 ligand
  • CD40 On normal cells, CD40 is expressed on cells with high proliferative potential, including hematopoietic progenitors, epithelial and endothelial cells, and all antigen-presenting cells (dendritic cells, activated B lymphocytes, and activated monocytes). CD40 is highly expressed on several types of B-cell hematologic malignancies including multiple myeloma, non-Hodgkin's lymphoma (NHL), and chronic lymphocytic leukemia (CLL). The high prevalence of CD40 expression on B-cell malignancies makes it an attractive potential tumor target for antibody-based cancer therapy. CD40 is also expressed on a majority of bladder cancers and a significant percentage of other solid tumors, including head and neck cancers, renal cell carcinomas, ovarian and lung cancer.
  • Anti-CD40 antibodies and their uses for treating B cell hematologic malignancies have been described. See, e.g., U.S. Pat. Nos. 6,946,129; 6,843,989; 6,838,261; WO 2000/075348; US-2002-0197256; WO 2006/128103; and WO 2007/075326. It has been shown that a humanized anti-CD40 antibody induces growth inhibition and apoptosis of CD40-positive cells in a subset of hematologic tumor cell lines through direct signal transduction. WO 2006/128103; WO 2007/075326.
  • the humanized anti-CD40 antibody kills tumor cells via immune effector functions, including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP).
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • SCID severe combined immunodeficient mice
  • anti-CD40 antibodies can induce growth inhibition and apoptosis of CD40-positive cells and may have anti-tumor activity in various types of B cell lymphoma patients, not all B lymphoma cells are sensitive to anti-CD40 antibody mediated cell death. There remains a need to identify one or more predictive markers for the responsiveness of B-cell lymphoma patients to anti-CD40 antibody therapy.
  • the invention provides methods and compositions for predicting, assessing or aiding assessment of responsiveness of a subject having a type of B-cell lymphoma to treatment with an anti-CD40 antibody.
  • the invention provides methods for assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to treatment with an anti-CD40 antibody, comprising comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7 and 13 in a B-cell lymphoma sample from the subject to a reference level.
  • the invention provides methods for predicting responsiveness or monitoring treatment/responsiveness to an anti-CD40 antibody treatment in a subject having a B-cell lymphoma, comprising comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7 and 13 in a B-cell lymphoma sample from the subject to a reference level.
  • the invention provides methods for predicting, assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to an anti-CD40 antibody treatment, comprising the steps of: (a) measuring expression level of one or more marker genes in a sample comprising B lymphoma cells obtained from said subject, wherein said one or more marker genes are selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7; (b) predicting whether the subject is likely to respond to the anti-CD40 antibody treatment based on the measured expression level of said one or more marker genes from step (a).
  • expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen maker genes from the group are measured and used for the prediction, assessment, or aiding assessment.
  • the prediction, assessment, or aiding assessment is determined by comparing the measured expression level of one or more marker genes to a reference level.
  • a reference level is a value or a range determined based on the measured expression level of the corresponding marker gene in samples comprising the B lymphoma cells from subjects having tumor volume increased or decreased after the anti-CD40 antibody treatment.
  • the invention provides methods preparing a personalized genomics profile for a subject having B-cell lymphoma comprising the steps of: (a) determining expression level of one or more marker genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, PUS7, and BCL6 in a sample comprising B lymphoma cells obtained from the subject; and (b) generating a report summarizing the expression level of one or more marker genes obtained in step (a).
  • expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or fifteen maker genes from the group are measured and used for the generating the report for the personalized genomics profile.
  • the report includes a recommendation for an anti-CD40 antibody treatment for the subject.
  • the recommendation is determined by comparing the measured expression level of the marker genes to a reference level.
  • a reference level is a value or a range determined based on the measured expression level of the corresponding marker gene in samples comprising the B lymphoma cells from subjects having tumor volume increased or decreased after the anti-CD40 antibody treatment.
  • the invention provides methods for predicting, assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to an anti-CD40 antibody treatment, comprising the steps of: (a) measuring expression level at least two marker genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 in a sample comprising B lymphoma cells from the subject; (b) calculating sensitivity index value (SI) based on the measured expression level of the marker genes in step (a) by the following equation:
  • ⁇ j is the coefficient value for each marker genes measured;
  • p is the number of marker genes measured;
  • x i is transformed, normalized expression level for the sample from the subject for expression level of each marker measured;
  • ⁇ j and ⁇ j are means and standard deviations for each marker gene measured; wherein ⁇ j , ⁇ j and ⁇ j are determined from patient samples comprising the B lymphoma cells.
  • a value equals or greater than zero for the sensitivity index indicates that the subject is likely to respond the anti-CD40 antibody treatment, or wherein a value less than zero for the sensitivity index indicates that the subject is less likely to respond the anti-CD40 antibody treatment.
  • the expression level of at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen marker genes are measured and used for the sensitivity index calculation.
  • the expression level of IFITM1, RGS13, CD79B, CD22, BTG2, CD44, EPDR1, and UAP1 are measured and used for the sensitivity index calculation.
  • the invention provides methods for treating a subject having a B-cell lymphoma, comprising administering an effective amount of the an anti-CD40 antibody to the subject, wherein the responsiveness of the B-cell lymphoma in the subject has been assessed by the methods described herein.
  • the invention provides methods for treating a subject having a B-cell lymphoma, comprising a) selecting a subject for an anti-CD40 antibody treatment by comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7 and 13 in a B-cell lymphoma sample from the subject to a reference level to assess if the B-cell lymphoma in the subject is suitable for the anti-CD40 antibody treatment; and administering an effective amount of the anti-CD40 antibody to the subject.
  • the reference level is a measured expression level of one or more reference genes in Table 8 or Table 9 in the B-cell lymphoma sample from the subject.
  • the reference level is a measured expression level of the marker gene in a different B-cell lymphoma sample.
  • the different B cell lymphoma sample comprises B lymphoma cells that are resistant to an anti-CD40 antibody induced cell death.
  • the measured expression level of the marker gene and/or the reference level are normalized.
  • measured expression levels of at least two, at least five, at least ten, at least fifteen, or at least twenty genes in any of Tables 2-4, 6, 7 and 13 in the B-cell lymphoma sample from the subject are compared to one or more reference levels.
  • the expression level is measured by detecting mRNA expression (e.g., real time quantitative reverse transcription PCR (qRT-PCR)) and/or by detecting protein expression (e.g., immunohistochemistry (IHC)).
  • mRNA expression e.g., real time quantitative reverse transcription PCR (qRT-PCR)
  • detecting protein expression e.g., immunohistochemistry (IHC)
  • the marker genes measured comprise one or more CD40 ligand downregulated genes (e.g., VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, and POU2AF1).
  • the marker genes measured comprise one or more genes in the B-cell receptor signaling pathway (e.g., CD22, RGS13, and MEF2B).
  • expression levels of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen genes selected from the group consisting of VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, and MEF2B in the B-cell lymphoma sample from the subject are compared to one or more reference levels.
  • expression levels of one or more gene pairs selected from the group consisting of VNN2 and EPDR1, RGS13 and EPDR1, CD22 and EPDR1, LRRC8A and PRPSAP2, CD40 and IGF1R, IFITM1 and BTG2, SMN1 and LMO2, PRKCA and YIPF3 in a the B-cell lymphoma sample are compared.
  • expression levels are compared between one or more gene pairs VNN2 and EPDR1, RGS13 and EPDR1, CD22 and EPDR1, LRRC8A and PRPSAP2, CD40 and IGF1R, IFITM1 and BTG2, SMN1 and LMO2, PRKCA and YIPF3 in the B-cell lymphoma sample, and sensitivity index calculated as the sum of signed t-scores for log 2-scale expression of the gene pairs is used to assess responsiveness of the B-cell lymphoma to an anti-CD40 antibody treatment.
  • the B-cell lymphoma is non-Hodgkin's lymphoma (NHL), including, but is not limited to, follicular lymphoma, relapsed follicular lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, lymphoplasmacytic lymphoma, mycosis fungoides/Sezary syndrome, splenic marginal zone lymphoma, and diffuse large B-cell lymphoma.
  • the B-cell lymphoma is selected from the group consisting of indolent lymphoma, aggressive lymphoma, and highly aggressive lymphoma.
  • kits comprising reagents for measuring expression levels of at least one marker gene in any of Tables 2-4, 6, 7 and 13.
  • the kits comprise at least a pair of primers for amplifying by PCR at least one marker gene in any of Tables 2-4, 6, 7 and 13.
  • forward and reverse primers shown in Table 10 may be used.
  • the kits may further comprise a pair of primers for amplifying a reference gene in Table 8.
  • the kits may further comprise a surface having attached thereof probes for detecting the amplified gene products, such as a microarray and the invention contemplates and includes such surfaces.
  • kits comprise at least a pair of primers and a probe for detecting expression level of one marker gene in any of Tables 2-4, 6, 7 and 13 by qRT-PCR.
  • the kits may further comprise a pair of primers and a probe for detecting expression level of a reference gene in Table 8 by qRT-PCR.
  • primer and probe sets shown in Table 10 may be used for detection expression level of genes by qRT-PCR.
  • the kits comprise one or more antibodies that specifically recognize one or more proteins encoded by the marker gene.
  • the kits may further comprise other reagents and/or instructions for carrying out any of the methods described herein.
  • FIG. 1 Enrichment plot of genes within BASSO_GERMINAL_CENTER_CD40_DN gene set.
  • the upper plot represents the enrichment score distribution across the ranked genes from the moderated t-test (Table 2).
  • the lower plot displays the distribution of the enrichment with respect to a ranked list metric known as signal2noice.
  • signal2noice a ranked list metric
  • FIG. 2 VNN2, a CD40L-downregulated gene, is overexpressed in sensitive NHL cells to anti-CD40 Ab.1 and discriminates between the two classes of sensitive and resistant.
  • the bar graph represents the mRNA expression level and the line graph represents the IC25 values.
  • FIG. 3A-3C RGS13, CD22, and MEF2B germinal center B markers, are overexpressed in sensitive and intermediate NHL cells to anti-CD40 Ab.1 and can discriminate with reasonable accuracy between the two classes of sensitive and resistant.
  • the bar graph represents the mRNA expression level and the line graph represents the IC25 values.
  • FIG. 4 Anti-CD40Ab.1 Sensitivity Index Scoring Across NHL Cell Lines. Stepwise Linear Modeling and gene-pair scoring was applied to each cell line based on mRNA expression data. The primary y-axis displays the anti-CD40 Ab.1 Sensitivity Index and the secondary y-axis displays the anti-CD40 Ab.1 IC25 values plotted against the NHL cell lines on the x-axis. A high anti-CD40 Ab.1 Sensitivity Index (> ⁇ 4) represents an increased probability of a cell line being sensitive.
  • FIG. 5 Correlation of CD40 signature genes with anti-CD40.Ab.1 sensitivity.
  • FIGS. 6-1 to 6 - 35 Gene bank sequences for genes listed in Table 7 and Table 10. Nucleic acid sequences encoding mRNA of VNN2 ( FIG. 6-1 : SEQ ID NO:258), RGS13 ( FIG. 6-2 : SEQ ID NO:259), CD22 ( FIGS. 6-3 and 6 - 4 : SEQ ID NO:260), LRRC8A ( FIG. 6-5 : SEQ ID NO:261), CD40 ( FIG. 6-6 : SEQ ID NO:262), IFITM1 ( FIG. 6-7 : SEQ ID NO:263), PRKCA ( FIGS. 6-8 to 6 - 10 : SEQ ID NO:264), BCL6 ( FIGS.
  • FIGS. 6-11 and 6 - 12 SEQ ID NO:265), EPDR1 ( FIG. 6-13 : SEQ ID NO:266), PRPSAP2 ( FIG. 6-14 : SEQ ID NO:267), IGF1R ( FIGS. 6-15 to 6 - 18 : SEQ ID NO:268), BTG2 ( FIGS. 6-19 and 6 - 20 : SEQ ID NO:269), LMO2 ( FIG. 6-21 : SEQ ID NO:270), YIPF3 ( FIG. 6-22 : SEQ ID NO:271), SMN1 ( FIG. 6-23 : SEQ ID NO:272), CD79B ( FIG. 6-24 : SEQ ID NO:273), CD44 ( FIGS.
  • FIG. 8 Association of BCL6 expression and percent change in SPD measurements for 26 patients with DLBCL. SPD percent change is determined by comparing the smallest post-baseline SPD to baseline SPD. Positive change indicates tumor volume increases, and negative change indicates tumor volume decreases.
  • the present invention is based on the discovery that certain genes (e.g., genes shown in Tables 2-4, 6, 7 and 13) are differentially expressed between B lymphoma cells that are sensitive to anti-CD40 antibody induced cell death and B lymphoma cells that are resistant to anti-CD40 induced cell death. Data from clinical trials described in Example 2 indicate that the expression level of the fourteen genes shown in Table 13 is highly associated with responsiveness to anti-CD40 Ab.1 treatment. Some of the differentially expressed genes between sensitive B lymphoma cells and resistant B lymphoma cells are the CD40 ligand downregulated pathway genes; and some are in the B-cell receptor signaling pathway.
  • expression levels of one or more of these differentially expressed genes can be used for assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to treatment with anti-CD40 antibodies, predicting responsiveness of the subject to treatment with anti-CD40 antibodies, and monitoring treatment/responsiveness in the subject.
  • Primers, oligonucleotides and polynucleotides employed in the present invention can be generated using standard techniques known in the art.
  • a subject having a B-cell lymphoma and “B-cell lymphoma patient” refer to a subject who has been diagnosed with a type of B-cell lymphoma or has been given a probable diagnosis of a type of B-cell lymphoma.
  • biomarker refers generally to a molecule, including a gene, protein, carbohydrate structure, or glycolipid, the expression of which in or on a mammalian tissue or cell or secreted can be detected by known methods (or methods disclosed herein) and is predictive or can be used to predict (or aid prediction) for a mammalian cell's or tissue's sensitivity to, and in some embodiments, to predict (or aid prediction) an individual's responsiveness to treatment regimes based on anti-CD40 antibodies.
  • sample refers to a composition that is obtained or derived from a subject of interest that contains a cellular and/or other molecular entity that is to be characterized and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics.
  • disease sample and variations thereof refers to any sample obtained from a subject of interest that would be expected or is known to contain the cellular and/or molecular entity that is to be characterized.
  • tissue or cell sample is meant a collection of similar cells obtained from a tissue of a subject or patient.
  • the source of the tissue or cell sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, or interstitial fluid; cells from any time in gestation or development of the subject.
  • the tissue sample may also be primary or cultured cells or cell lines.
  • the tissue or cell sample is obtained from a disease tissue/organ.
  • the tissue sample may contain compounds which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
  • a “section” of a tissue sample is meant a single part or piece of a tissue sample, e.g. a thin slice of tissue or cells cut from a tissue sample. It is understood that multiple sections of tissue samples may be taken and subjected to analysis according to the present invention, provided that it is understood that the present invention comprises a method whereby the same section of tissue sample is analyzed at both morphological and molecular levels, or is analyzed with respect to both protein and nucleic acid.
  • a “B-cell lymphoma sample” or a “sample comprising B lymphoma cells” is a tissue or cell sample containing B lymphoma cells from a subject or a patient that have been diagnosed with a type of B-cell lymphoma.
  • method for “aiding assessment” refers to methods that assist in making a clinical determination (e.g., responsiveness of a B-cell lymphoma to treatment with anti-CD40 antibodies), and may or may not be conclusive with respect to the definitive assessment.
  • a “subject” or an “individual” is a mammal, more preferably a human. Mammals include, but are not limited to, humans, primates, farm animal, sport animals, rodents, and pets (e.g., dogs and cats).
  • a “reference value” can be an absolute value; a relative value; a value that has an upper and/or lower limit; a range of values; an average value; a median value; a mean value; or a value as compared to a particular control or baseline value.
  • array refers to an ordered arrangement of hybridizable array elements, such as polynucleotide probes (e.g., oligonucleotides) and antibodies, on a substrate.
  • the substrate can be a solid substrate, such as a glass slide, or a semi-solid substrate, such as nitrocellulose membrane.
  • the nucleotide sequences can be DNA, RNA, or any permutations thereof.
  • “Amplification,” as used herein, generally refers to the process of producing multiple copies of a desired sequence. “Multiple copies” means at least 2 copies. A “copy” does not necessarily mean perfect sequence complementarity or identity to the template sequence. For example, copies can include nucleotide analogs such as deoxyinosine, intentional sequence alterations (such as sequence alterations introduced through a primer comprising a sequence that is hybridizable, but not complementary, to the template), and/or sequence errors that occur during amplification.
  • Expression/amount of a gene or biomarker in a first sample is at a level “greater than” the level in a second sample if the expression level/amount of the gene or biomarker in the first sample is at least about 1.5 ⁇ , 1.75 ⁇ , 2 ⁇ , 3 ⁇ , 4 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ or 10 ⁇ the expression level/amount of the gene or biomarker in the second sample.
  • Expression levels/amounts can be determined based on any suitable criterion known in the art, including but not limited to mRNA, cDNA, proteins, protein fragments and/or gene copy. Expression levels/amounts can be determined qualitatively and/or quantitatively.
  • Polynucleotide or “nucleic acid,” as used interchangeably herein, refer to polymers of nucleotides of any length, and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the polymer.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • modifications include, for example, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, cabamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing al
  • any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports.
  • the 5′ and 3′ terminal OH can be phosphorylated or substituted with amines or organic capping groups moieties of from 1 to 20 carbon atoms.
  • Other hydroxyls may also be derivatized to standard protecting groups.
  • Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2′-O-methyl-2′-O-allyl, 2′-fluoro- or 2′-azido-ribose, carbocyclic sugar analogs, ⁇ -anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and a basic nucleoside analogs such as methyl riboside.
  • One or more phosphodiester linkages may be replaced by alternative linking groups.
  • linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S(“thioate”), P(S)S (“dithioate”), “(O)NR 2 (“amidate”), P(O)R, P(O)OR′, CO or CH 2 (“formacetal”), in which each R or R′ is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (—O—) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • Oligonucleotide generally refers to short, generally single stranded, generally synthetic polynucleotides that are generally, but not necessarily, less than about 200 nucleotides in length.
  • oligonucleotide and polynucleotide are not mutually exclusive. The description above for polynucleotides is equally and fully applicable to oligonucleotides.
  • a “primer” is generally a short single stranded polynucleotide, generally with a free 3′-OH group, that binds to a target potentially present in a sample of interest by hybridizing with a target sequence, and thereafter promotes polymerization of a polynucleotide complementary to the target.
  • a “pair of primers” refer to a 5′ primer and a 3′ primer that can be used to amplify a portion of a specific target gene.
  • 3′ generally refers to a region or position in a polynucleotide or oligonucleotide 3′ (downstream) from another region or position in the same polynucleotide or oligonucleotide.
  • 5′ generally refers to a region or position in a polynucleotide or oligonucleotide 5′ (upstream) from another region or position in the same polynucleotide or oligonucleotide.
  • gene amplification refers to a process by which multiple copies of a gene or gene fragment are formed in a particular cell or cell line.
  • the duplicated region (a stretch of amplified DNA) is often referred to as “amplicon.”
  • amplicon a stretch of amplified DNA
  • the amount of the messenger RNA (mRNA) produced i.e., the level of gene expression, also increases in the proportion of the number of copies made of the particular gene expressed.
  • Detection includes any means of detecting, including direct and indirect detection.
  • prediction is used herein to refer to the likelihood that a patient will respond either favorably or unfavorably to a drug or set of drugs. In one embodiment, the prediction relates to the extent of those responses. In one embodiment, the prediction relates to whether and/or the probability that a patient will survive or improve following treatment, for example treatment with a particular therapeutic agent, and for a certain period of time without disease recurrence.
  • the predictive methods of the invention can be used clinically to make treatment decisions by choosing the most appropriate treatment modalities for any particular patient.
  • the predictive methods of the present invention are valuable tools in predicting if a patient is likely to respond favorably to a treatment regimen, such as a given therapeutic regimen, including for example, administration of a given therapeutic agent or combination, surgical intervention, steroid treatment, etc., or whether long-term survival of the patient, following a therapeutic regimen is likely.
  • a treatment regimen such as a given therapeutic regimen, including for example, administration of a given therapeutic agent or combination, surgical intervention, steroid treatment, etc., or whether long-term survival of the patient, following a therapeutic regimen is likely.
  • long-term survival is used herein to refer to survival for at least 1 year, 5 years, 8 years, or 10 years following therapeutic treatment.
  • “Patient response” can be assessed using any endpoint indicating a benefit to the patient, including, without limitation, (1) inhibition, to some extent, of disease progression, including slowing down and complete arrest; (2) reduction in the number of disease episodes and/or symptoms; (3) reduction in lesional size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of disease cell infiltration into adjacent peripheral organs and/or tissues; (5) inhibition (i.e. reduction, slowing down or complete stopping) of disease spread; (6) relief, to some extent, of one or more symptoms associated with the disorder; (7) increase in the length of disease-free presentation following treatment; and/or (8) decreased mortality at a given point of time following treatment.
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonal antibodies), multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity or function.
  • Antibody fragments comprise a portion of a full length antibody, generally the antigen binding or variable region thereof.
  • Examples of antibody fragments include Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • “Fv” is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. From the folding of these two domains emanate six hypervariable loops (3 loops each from the H and L chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • the term “monoclonal antibody” as used herein refers to an antibody from a population of substantially homogeneous antibodies, i e., the individual antibodies comprising the population are identical and/or bind the same epitope(s), except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • Such monoclonal antibody typically includes an antibody comprising a polypeptide sequence that binds a target, wherein the target-binding polypeptide sequence was obtained by a process that includes the selection of a single target binding polypeptide sequence from a plurality of polypeptide sequences.
  • the selection process can be the selection of a unique clone from a plurality of clones, such as a pool of hybridoma clones, phage clones or recombinant DNA clones.
  • the selected target binding sequence can be further altered, for example, to improve affinity for the target, to humanize the target binding sequence, to improve its production in cell culture, to reduce its immunogenicity in vivo, to create a multispecific antibody, etc., and that an antibody comprising the altered target binding sequence is also a monoclonal antibody of this invention.
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the monoclonal antibody preparations are advantageous in that they are typically uncontaminated by other immunoglobulins.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including, for example, the hybridoma method (e.g., Kohler et al., Nature, 256:495 (1975); Harlow et al., Antibodies: A Laboratory Manual , (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling et al., in: Monoclonal Antibodies and T - Cell Hybridomas 563-681, (Elsevier, N.Y., 1981)), recombinant DNA methods (see, e.g., U.S. Pat. No.
  • phage display technologies see, e.g., Clackson et al., Nature, 352:624-628 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991); Sidhu et al., J. Mol. Biol. 338(2):299-310 (2004); Lee et al., J. Mol. Biol. 340(5):1073-1093 (2004); Fellouse, Proc. Nat. Acad. Sci. USA 101(34):12467-12472 (2004); and Lee et al. J. Immunol.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies.
  • “Chimeric” antibodies immunoglobulins have a portion of the heavy and/or light chain identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984)).
  • Humanized antibody as used herein is a subset of chimeric antibodies.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient or acceptor antibody) in which hypervariable region residues of the recipient are replaced by hypervariable region residues from a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues which are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance such as binding affinity.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence although the FR regions may include one or more amino acid substitutions that improve binding affinity.
  • the number of these amino acid substitutions in the FR are typically no more than 6 in the H chain, and in the L chain, no more than 3.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • a “human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human and/or has been made using any of the known techniques for making human antibodies. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • affinity matured antibody is one with one or more alterations in one or more CDRs/HVRs thereof which result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s).
  • Preferred affinity matured antibodies will have nanomolar or even picomolar affinities for the target antigen.
  • Affinity matured antibodies are produced by procedures known in the art. Marks et al. Bio/Technology 10:779-783 (1992) describes affinity maturation by VH and VL domain shuffling. Random mutagenesis of CDR/HVR and/or framework residues is described by: Barbas et al. Proc Nat. Acad.
  • Fc region is used to define the C-terminal region of an immunoglobulin heavy chain which may be generated by papain digestion of an intact antibody.
  • the Fc region may be a native sequence Fc region or a variant Fc region.
  • the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at about position Cys226, or from about position Pro230, to the carboxyl-terminus of the Fc region.
  • the Fc region of an immunoglobulin generally comprises two constant domains, a CH2 domain and a CH3 domain, and optionally comprises a CH4 domain.
  • Fc region chain herein is meant one of the two polypeptide chains of an Fc region.
  • Antibody effector functions refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody, and vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs Fc receptors
  • cytotoxic cells e.g. Natural Killer (NK) cells, neutrophils, and macrophages
  • NK cells Natural Killer cells
  • neutrophils neutrophils
  • macrophages cytotoxic cells
  • the antibodies “arm” the cytotoxic cells and are absolutely required for such killing.
  • the primary cells for mediating ADCC, NK cells express Fc ⁇ RIII only, whereas monocytes express Fc ⁇ RI, Fc ⁇ RII and Fc ⁇ RIII.
  • ADCC activity of a molecule of interest is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991).
  • an in vitro ADCC assay such as that described in U.S. Pat. No. 5,500,362 or 5,821,337 or Presta U.S. Pat. No. 6,737,056 may be performed.
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. PNAS ( USA ) 95:652-656 (1998).
  • Treating” or “treatment” or “alleviation” refers to therapeutic treatment wherein the object is to slow down (lessen) if not cure the targeted pathologic condition or disorder or prevent recurrence of the condition.
  • a subject is successfully “treated” for the B cell malignancy if, after receiving a therapeutic amount of a CD40 binding antibody, the subject shows observable and/or measurable reduction in or absence of one or more signs and symptoms of the particular disease.
  • the antibodies of the invention achieve >95% peripheral blood B cell depletion and the B cells return to 25% of baseline.
  • treatment with the anti-CD40 antibodies is effective to result in the cancer patients being progression-free in the cancer 3 months after treatment, preferably 6 months, more preferably one year, even more preferably 2 or more years post treatment.
  • non-Hodgkin's lymphoma refers to a cancer of the lymphatic system other than Hodgkin's lymphomas.
  • Hodgkin's lymphomas can generally be distinguished from non-Hodgkin's lymphomas by the presence of Reed-Sternberg cells in Hodgkin's lymphomas and the absence of said cells in non-Hodgkin's lymphomas.
  • an “effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • a “therapeutically effective amount” of a therapeutic agent may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the therapeutic agent are outweighed by the therapeutically beneficial effects.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • housekeeping gene refers to a group of genes that codes for proteins whose activities are essential for the maintenance of cell function. These genes are typically similarly expressed in all cell types.
  • correlate or “correlating” is meant comparing, in any way, the performance and/or results of a first analysis or protocol with the performance and/or results of a second analysis or protocol. For example, one may use the results of a first analysis or protocol in carrying out a second protocols and/or one may use the results of a first analysis or protocol to determine whether a second analysis or protocol should be performed. With respect to the embodiment of gene expression analysis or protocol, one may use the results of the gene expression analysis or protocol to determine whether a specific therapeutic regimen should be performed.
  • label when used herein refers to a compound or composition which is conjugated or fused directly or indirectly to a reagent such as a nucleic acid probe or an antibody and facilitates detection of the reagent to which it is conjugated or fused.
  • the label may itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • the invention provides methods for assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to treatment with an anti-CD40 antibody.
  • the invention also provides methods for predicting responsiveness or monitoring treatment/responsiveness to an anti-CD40 antibody treatment in a subject having a B-cell lymphoma.
  • the invention provides methods for selecting a subject having a B-cell lymphoma suitable for treatment with an anti-CD40 antibody and following up with an anti-CD40 antibody treatment.
  • the methods comprise measuring expression level of one or more marker genes in any of Tables 2-4, 6, 7, and 13 in a sample comprising B lymphoma cells obtained from the subject; and predicting, assessing, or aiding assessment of responsiveness of the subject to an anti-CD40 antibody treatment based on the measure expression level of said one or more marker genes.
  • the methods comprise comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7, and 13 in a B-cell lymphoma sample from the subject to a reference level for the respective marker gene.
  • the methods of the present invention are useful for clinicians to identify patients with B-cell lymphoma for treatment with an anti-CD40 antibody, aiding in patient selection during the course of development of anti-CD40 antibody therapy, prediction of likelihood of success when treating an individual patient with a particular treatment regimen, in assessing and monitoring disease progression, in monitoring treatment efficacy, and in determining prognosis for individual patients. Any of these embodiments are included in this invention.
  • the B-cell lymphoma is non-Hodgkin's lymphoma (NHL), including, but is not limited to, follicular lymphoma, relapsed follicular lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, lymphoplasmacytic lymphoma, mycosis fungoides/Sezary syndrome, splenic marginal zone lymphoma, and diffuse large B-cell lymphoma.
  • NHL Hodgkin's lymphoma
  • the B-cell lymphoma is indolent. In some embodiments, the B-cell lymphoma is aggressive. In some embodiments, the B-cell lymphoma is highly aggressive. In some embodiments, the indolent B-cell lymphoma is follicular lymphoma, marginal zone lymphoma, or small lymphocytic lymphoma. In some embodiments, the indolent B-cell lymphoma is follicular lymphoma.
  • the expression level of one or more of the marker genes in a B-cell lymphoma sample relative a reference level may be used in the methods of the invention, such as to predict, assess or aid assessment of responsiveness of the B-cell lymphoma to treatment with an anti-CD40 antibody.
  • Anti-CD40 antibody sensitive cells are cells having an IC25 value less than 0.4 ⁇ g/ml in reduction of cell viability by an anti-CD40 antibody tested as described in Example 1.
  • Anti-CD40 resistant cells are cells having an IC25 value greater than 1 ⁇ g/ml in reduction in cell viability as tested in Example 1.
  • genes in Tables 2-4, 6 and 7 are in the CD40 ligand downregulated pathway (for example, VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, and POU2AF1); and some of the genes in the tables are in the B-cell receptor signaling pathway (for example, CD22, RGS13, and MEF2B). Further, association of the expression level of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 (Table 13) has been confirmed by clinical trials described in Example 2.
  • CD40 ligand downregulated pathway for example, VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, and POU2AF1
  • B-cell receptor signaling pathway for example, CD22, RGS13, and
  • Expression levels of one or more of these genes are used in the methods of the invention. In some embodiments, expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least twenty, at least twenty five, or at least thirty genes are used in the methods of the invention.
  • expression levels of one or more of genes selected from the group consisting of VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, and MEF2B are measured and/or used.
  • expression levels of one or more of genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are measured and/or used.
  • expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen of these genes are measured and/or used.
  • expression levels of CD22, CD40, and BCL6 are measured and/or used.
  • expression levels of CD40, RGS13, CD22, BTG2, IGF1R, and CD44 are measured and/or used.
  • expression levels of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are measured and/or used.
  • expression levels of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or fifteen of genes in Table 7 or Table 13 are measured and/or used.
  • Genes (including sequences) identified in Tables 2-4, 6, 7 and 13 are known in the art.
  • the examples of Gene Bank accession numbers for human genes are VNN2 (NM — 004665; NM — 078488; AJ132100; D89974; BC064641; CR609799; BC126145; BC126147; and AB026705); RGS13 (NM — 002927; NM — 144766; BT006929; BC056866; AY562947; CR536532; CR610389; CR599001; BC016667; AF493935; BC036950; and AF030107); CD22 (NM — 001771; AK026467; BC109306; BC109307; AK225694; AK225625; X52785; and X59350); LRRC8A (AY143166; BC051322; AK123611; AY358286; NM — 01
  • the measured expression level of one or more marker genes in a B-cell lymphoma sample is compared to a reference level.
  • the reference level is the expression level of a gene the expression level of which does not change (does not change significantly) among different type of B-cell lymphomas, for example, between B-cell lymphoma sensitive to anti-CD40 antibody and B-cell lymphoma resistant to anti-CD40 antibody.
  • expression levels of one or more housekeeping genes shown in Table 8 are used as reference levels.
  • expression levels of one or more housekeeping genes shown in Table 9 are used as reference levels.
  • the measured expression level of the marker gene is normalized using the reference level. In some embodiments, the normalized expression level of the marker gene is calculated as a ratio of or difference between the marker gene and reference expression levels, on the original or on a log scale, respectively.
  • the reference genes in Table 8 and Table 9 were selected as specific normalizing counterparts to the marker genes in Table 4. Reference genes were selected for high mean expression and low variance in B cell lymphoma samples. In addition, reference genes were selected to have similar variance between replicated expression measurements of individual cell lines relative to variance between expression measurements of biologically distinct cell lines. In addition, reference genes were selected to have low statistical association with one or more markers in Table 4.
  • the reference level is a measured expression level of the marker gene in a different B-cell lymphoma sample.
  • the different B cell lymphoma sample comprises B lymphoma cells that are resistant to an anti-CD40 antibody induced cell death.
  • the reference level is determined based on the expression level of the corresponding marker gene in samples comprising B lymphoma cells from subjects having tumor volume increased after the anti-CD40 antibody treatment and/or having tumor volume decreased after the anti-CD40 antibody treatment.
  • the samples from subjects for reference level determination comprise the same type of B lymphoma cells as the sample from the subject whose responsiveness to the anti-CD40 antibody treatment is predicted or assessed.
  • the same method e.g., qRT-PCR
  • reagents e.g., primers and probes
  • VarW. vscr. SCR. IC25. GCB. SCR EXT. Probe symb.gse VarB mean var vscr rank P.
  • the methods disclosed herein provide methods to examine expression level of one or more of these marker genes in a lymphoma sample (e.g., B-cell lymphoma sample) relative a reference level.
  • the methods and assays include those which examine expression of marker genes such as one or more of those listed in any of Tables 2-4, 6, 7 and 13. Expression levels may be measured at mRNA level and/or protein level.
  • the invention provides methods for measuring levels of expression from a mammalian tissue or cells sample (such as cells and/or tissues associated with B-cell lymphoma). For example, for obtaining patient samples, H&E staining is carried out and used as a guide for tissue macrodissection to enrich for tumor content.
  • the sample can be obtained by a variety of procedures known in the art including, but is not limited to surgical excision, aspiration or biopsy.
  • the sample may be fresh or frozen.
  • the sample is fixed and embedded in paraffin or the like.
  • a mammalian tissue or cell sample is obtained and examined for expression of one or more biomarkers.
  • the methods may be conducted in a variety of assay formats, including assays detecting mRNA expression, enzymatic assays detecting presence of enzymatic activity, and immunohistochemistry assays. Determination of expression of such biomarkers in said tissues or cells will be predictive that such tissues or cells will be sensitive/responsive to treatment with an anti-CD40 antibody.
  • expression of various biomarkers in a sample can be analyzed by a number of methodologies, many of which are known in the art and understood by the skilled artisan, including but not limited to, microarray (gene and/or tissue array analysis), in situ hybridization, Northern analysis, PCR analysis of mRNAs, immunohistochemical and/or Western analysis, quantitative blood based assays (as for example Serum ELISA) (to examine, for example, levels of protein expression), and/or biochemical enzymatic activity assays.
  • Typical protocols for evaluating the status of genes and gene products are found, for example in Ausubel et al.
  • the methods of the invention further include protocols which examine the presence and/or expression of mRNAs, such as mRNAs of genes listed in any of Tables 2-4, 6, 7 and 13, in a tissue or cell sample.
  • expression of various biomarkers in a sample may be analyzed by microarray technologies, which examine or detect mRNAs, such as mRNAs in any of Tables 2-4, 6, 7 and 13, in a tissue or cell sample.
  • test and control mRNA samples from test and control tissue samples are reverse transcribed and labeled to generate cDNA probes. The probes are then hybridized to an array of nucleic acids immobilized on a solid support.
  • the array is configured such that the sequence and position of each member of the array is known. For example, a selection of genes that have potential to be expressed in certain disease states may be arrayed on a solid support. Hybridization of a labeled probe with a particular array member indicates that the sample from which the probe was derived expresses that gene. Differential gene expression analysis of disease tissue can provide valuable information. Microarray technology utilizes nucleic acid hybridization techniques and computing technology to evaluate the mRNA expression profile of thousands of genes within a single experiment. (See, e.g., WO 01/75166 published Oct. 11, 2001; see also, for example, U.S. Pat. Nos.
  • DNA microarrays are miniature arrays containing gene fragments that are either synthesized directly onto or spotted onto glass or other substrates. Thousands of genes are usually represented in a single array.
  • a typical microarray experiment involves the following steps: 1) preparation of fluorescently labeled target from RNA isolated from the sample, 2) hybridization of the labeled target to the microarray, 3) washing, staining, and scanning of the array, 4) analysis of the scanned image and 5) generation of gene expression profiles.
  • oligonucleotide usually 25 to 70 mers
  • gene expression arrays containing PCR products prepared from cDNAs can be either prefabricated and spotted to the surface or directly synthesized on to the surface (in situ).
  • the Affymetrix GeneChip® system is a commercially available microarray system which comprises arrays fabricated by direct synthesis of oligonucleotides on a glass surface.
  • Probe/Gene Arrays Oligonucleotides, usually 25 mers, are directly synthesized onto a glass wafer by a combination of semiconductor-based photolithography and solid phase chemical synthesis technologies. Each array contains up to 400,000 different oligos and each oligo is present in millions of copies. Since oligonucleotide probes are synthesized in known locations on the array, the hybridization patterns and signal intensities can be interpreted in terms of gene identity and relative expression levels by the Affymetrix Microarray Suite software.
  • Each gene is represented on the array by a series of different oligonucleotide probes.
  • Each probe pair consists of a perfect match oligonucleotide and a mismatch oligonucleotide.
  • the perfect match probe has a sequence exactly complimentary to the particular gene and thus measures the expression of the gene.
  • the mismatch probe differs from the perfect match probe by a single base substitution at the center base position, disturbing the binding of the target gene transcript. This helps to determine the background and nonspecific hybridization that contributes to the signal measured for the perfect match oligo.
  • the Microarray Suite software subtracts the hybridization intensities of the mismatch probes from those of the perfect match probes to determine the absolute or specific intensity value for each probe set.
  • Probes are chosen based on current information from GenBank and other nucleotide repositories. The sequences are believed to recognize unique regions of the 3′ end of the gene.
  • a GeneChip Hybridization Oven (“rotisserie” oven) is used to carry out the hybridization of up to 64 arrays at one time.
  • the fluidics station performs washing and staining of the probe arrays. It is completely automated and contains four modules, with each module holding one probe array. Each module is controlled independently through Microarray Suite software using preprogrammed fluidics protocols.
  • the scanner is a confocal laser fluorescence scanner which measures fluorescence intensity emitted by the labeled cRNA bound to the probe arrays.
  • the computer workstation with Microarray Suite software controls the fluidics station and the scanner.
  • Microarray Suite software can control up to eight fluidics stations using preprogrammed hybridization, wash, and stain protocols for the probe array.
  • the software also acquires and converts hybridization intensity data into a presence/absence call for each gene using appropriate algorithms.
  • the software detects changes in gene expression between experiments by comparison analysis and formats the output into .txt files, which can be used with other software programs for further data analysis.
  • expression of various biomarkers in a sample may also be assessed by examining gene deletion or gene amplification.
  • Gene deletion or amplification may be measured by any one of a wide variety of protocols known in the art, for example, by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA (Thomas, Proc. Natl. Acad. Sci. USA, 77:5201-5205 (1980)), dot blotting (DNA analysis), or in situ hybridization (e.g., FISH), using an appropriately labeled probe, cytogenetic methods or comparative genomic hybridization (CGH) using an appropriately labeled probe.
  • these methods may be employed to detect deletion or amplification of genes listed in any of Tables 2-4, 6, 7 and 13.
  • expression of various biomarkers in a sample may be assessed by hybridization assays using complementary DNA probes (such as in situ hybridization using labeled riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers, such as primers specific for one or more genes listed in any of Tables 2-4, 6, 7 and 13, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like).
  • complementary DNA probes such as in situ hybridization using labeled riboprobes, Northern blot and related techniques
  • nucleic acid amplification assays such as RT-PCR using complementary primers, such as primers specific for one or more genes listed in any of Tables 2-4, 6, 7 and 13, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like.
  • Tissue or cell samples from mammals can be conveniently assayed for, e.g., mRNAs of genes listed in any of Tables 2-4, 6, 7 and 13, using Northern, dot blot or PCR analysis.
  • expression of one or more biomarkers may be assayed by RT-PCR.
  • the RT-PCR may be quantitative RT-PCR (qRT-PCR).
  • the RT-PCR is real-time RT-PCR.
  • the RT-PCR is quantitative real-time RT-PCR.
  • RT-PCR assays such as quantitative PCR assays are well known in the art.
  • a method for detecting a mRNA in a biological sample comprises producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using a polynucleotide as sense and antisense primers to amplify cDNAs therein; and detecting the presence of the amplified cDNA of interest.
  • the real-time RT-PCR may be quantitative RT-PCR.
  • the real-time RT-PCR may be performed using TaqMan® chemistry (Applied Biosystems).
  • the real-time RT-PCR may be performed using TaqMan® chemistry (Applied Biosystems) and the ABI Prism® 7700 Sequence Detection System (Applied Biosystems).
  • the real-time RT-PCR combines the principles that Taq polymerase has a 5′-3; exonuclease activity and dual-labeled fluorogenic oligonucleotide problems have been created which emit a fluorescent signal only upon cleavage, based on the principle of fluorescence resonance energy transfer. See, e.g., Overbergh, L. et al., J. Biomolecular Techniques 14(1): 33-43 (2003).
  • such methods can include one or more steps that allow one to determine the levels of mRNA, such as a mRNA of genes listed in any of Tables 2-4, 6, 7 and 13, in a biological sample (e.g., by simultaneously examining the levels a comparative control mRNA sequence of a “housekeeping” gene such as an actin family member and/or one or more genes listed in Table 8 or Table 9).
  • mRNA such as a mRNA of genes listed in any of Tables 2-4, 6, 7 and 13, in a biological sample
  • a comparative control mRNA sequence of a “housekeeping” gene such as an actin family member and/or one or more genes listed in Table 8 or Table 9.
  • the expression of proteins encoded by the genes listed in any of Tables 2-4, 6, 7 and 13 in a sample is examined using immunohistochemistry and staining protocols.
  • Immunohistochemical staining of tissue sections has been shown to be a reliable method of assessing or detecting presence of proteins in a sample.
  • Immunohistochemistry (“IHC”) techniques utilize an antibody to probe and visualize cellular antigens in situ, generally by chromogenic or fluorescent methods.
  • tissue or cell sample from a mammal may be used.
  • samples include, but are not limited to, tissue biopsy, blood, lung aspirate, sputum, lymph fluid, etc.
  • the sample can be obtained by a variety of procedures known in the art including, but not limited to surgical excision, aspiration or biopsy.
  • the tissue may be fresh or frozen.
  • the sample is fixed and embedded in paraffin or the like.
  • the tissue sample may be fixed (i.e. preserved) by conventional methodology (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology,” 3 rd edition (1960) Lee G. Luna, H T (ASCP) Editor, The Blakston Division McGraw-Hill Book Company, New York; The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology (1994) Ulreka V. Mikel, Editor, Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C.).
  • a fixative is determined by the purpose for which the sample is to be histologically stained or otherwise analyzed.
  • the length of fixation depends upon the size of the tissue sample and the fixative used.
  • neutral buffered formalin, Bouin's or paraformaldehyde may be used to fix a sample.
  • the sample is first fixed and is then dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned. Alternatively, one may section the tissue and fix the sections obtained.
  • the tissue sample may be embedded and processed in paraffin by conventional methodology (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology”, supra).
  • paraffin that may be used include, but are not limited to, Paraplast, Broloid, and Tissuemay.
  • the sample may be sectioned by a microtome or the like (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology”, supra). By way of example for this procedure, sections may range from about three microns to about five microns in thickness.
  • the sections may be attached to slides by several standard methods. Examples of slide adhesives include, but are not limited to, silane, gelatin, poly-L-lysine and the like.
  • the paraffin embedded sections may be attached to positively charged slides and/or slides coated with poly-L-lysine.
  • the tissue sections are generally deparaffinized and rehydrated to water.
  • the tissue sections may be deparaffinized by several conventional standard methodologies. For example, xylenes and a gradually descending series of alcohols may be used (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology”, supra).
  • commercially available deparaffinizing non-organic agents such as Hemo-De7 (CMS, Houston, Tex.) may be used.
  • a tissue section may be analyzed using IHC.
  • IHC may be performed in combination with additional techniques such as morphological staining and/or fluorescence in-situ hybridization.
  • Two general methods of IHC are available; direct and indirect assays. According to the first assay, binding of antibody to the target antigen (e.g., a protein or fragment thereof encoded by one or more genes listed in Tables 1-4, 6 and 7) is determined directly.
  • This direct assay uses a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction.
  • unconjugated primary antibody binds to the antigen and then a labeled secondary antibody binds to the primary antibody.
  • a labeled secondary antibody binds to the primary antibody.
  • a chromogenic or fluorogenic substrate is added to provide visualization of the antigen. Signal amplification occurs because several secondary antibodies may react with different epitopes on the primary antibody.
  • the primary and/or secondary antibody used for immunohistochemistry typically will be labeled with a detectable moiety.
  • Numerous labels are available which can be generally grouped into the following categories:
  • Radioisotopes such as 35 S, 14 C, 125 I, 3 H, and 131 I.
  • the antibody can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology , Volumes 1 and 2, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
  • Fluorescent labels including, but are not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE7 and SPECTRUM GREEN7 and/or derivatives of any one or more of the above.
  • the fluorescent labels can be conjugated to the antibody using the techniques disclosed in Current Protocols in Immunology , supra, for example. Fluorescence can be quantified using a fluorimeter.
  • the enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, ⁇ -galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • luciferases e.g., firefly luciferase and bacterial lucifera
  • enzyme-substrate combinations include, for example:
  • HRPO Horseradish peroxidase
  • HPO horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3′,5,5′-tetramethyl benzidine hydrochloride
  • ⁇ -D-galactosidase ( ⁇ -D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl-1′-D-galactosidase) or fluorogenic substrate (e.g., 4-methylumbelliferyl- ⁇ -D-galactosidase).
  • a chromogenic substrate e.g., p-nitrophenyl-1′-D-galactosidase
  • fluorogenic substrate e.g., 4-methylumbelliferyl- ⁇ -D-galactosidase
  • the label is indirectly conjugated with the antibody.
  • the antibody can be conjugated with biotin and any of the four broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner.
  • the antibody is conjugated with a small hapten and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody.
  • indirect conjugation of the label with the antibody can be achieved.
  • tissue section prior to, during or following IHC may be desired.
  • epitope retrieval methods such as heating the tissue sample in citrate buffer may be carried out (see, e.g., Leong et al. Appl. Immunohistochem. 4(3):201 (1996)).
  • the tissue section is exposed to primary antibody for a sufficient period of time and under suitable conditions such that the primary antibody binds to the target protein antigen in the tissue sample.
  • Appropriate conditions for achieving this can be determined by routine experimentation.
  • the extent of binding of antibody to the sample is determined by using any one of the detectable labels discussed above.
  • the label is an enzymatic label (e.g. HRPO) which catalyzes a chemical alteration of the chromogenic substrate such as 3,3′-diaminobenzidine chromogen.
  • the enzymatic label is conjugated to antibody which binds specifically to the primary antibody (e.g. the primary antibody is rabbit polyclonal antibody and secondary antibody is goat anti-rabbit antibody).
  • the antibodies employed in the IHC analysis to detect expression of one or more biomarkers are antibodies generated to bind primarily to the one or more biomarkers of interest, such as one or more proteins encoded by genes listed in any of Tables 2-4, 6 and 7.
  • the antibody is a monoclonal antibody. Antibodies are readily available in the art, including from various commercial sources, and can also be generated using routine skills known in the art.
  • staining intensity criteria may be evaluated as follows:
  • the sample may be contacted with an antibody specific for said biomarker under conditions sufficient for an antibody-biomarker complex to form, and then detecting said complex.
  • the presence of the biomarker may be detected in a number of ways, such as by Western blotting and ELISA procedures for assaying a wide variety of tissues and samples, including plasma or serum.
  • a wide range of immunoassay techniques using such an assay format are available, see, e.g., U.S. Pat. Nos. 4,016,043, 4,424,279 and 4,018,653. These include both single-site and two-site or “sandwich” assays of the non-competitive types, as well as in the traditional competitive binding assays. These assays also include direct binding of a labeled antibody to a target biomarker.
  • Sandwich assays are among the most useful and commonly used assays. A number of variations of the sandwich assay technique exist, and all are intended to be encompassed by the present invention. Briefly, in a typical forward assay, an unlabelled antibody is immobilized on a solid substrate, and the sample to be tested brought into contact with the bound molecule. After a suitable period of incubation, for a period of time sufficient to allow formation of an antibody-antigen complex, a second antibody specific to the antigen, labeled with a reporter molecule capable of producing a detectable signal is then added and incubated, allowing time sufficient for the formation of another complex of antibody-antigen-labeled antibody.
  • any unreacted material is washed away, and the presence of the antigen is determined by observation of a signal produced by the reporter molecule.
  • the results may either be qualitative, by simple observation of the visible signal, or may be quantitated by comparing with a control sample containing known amounts of biomarker.
  • a simultaneous assay in which both sample and labeled antibody are added simultaneously to the bound antibody.
  • a first antibody having specificity for the biomarker is either covalently or passively bound to a solid surface.
  • the solid surface is typically glass or a polymer, the most commonly used polymers being cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • the solid supports may be in the form of tubes, beads, discs of microplates, or any other surface suitable for conducting an immunoassay.
  • the binding processes are well-known in the art and generally consist of cross-linking covalently binding or physically adsorbing, the polymer-antibody complex is washed in preparation for the test sample. An aliquot of the sample to be tested is then added to the solid phase complex and incubated for a period of time sufficient (e.g., 2-40 minutes or overnight if more convenient) and under suitable conditions (e.g., from room temperature to 40° C. such as between 25° C. and 32° C. inclusive) to allow binding of any subunit present in the antibody. Following the incubation period, the antibody subunit solid phase is washed and dried and incubated with a second antibody specific for a portion of the biomarker. The second antibody is linked to a reporter molecule which is used to indicate the binding of the second antibody to the molecular marker.
  • the methods involves immobilizing the target biomarkers in the sample and then exposing the immobilized target to specific antibody which may or may not be labeled with a reporter molecule.
  • a bound target may be detectable by direct labeling with the antibody.
  • a second labeled antibody, specific to the first antibody is exposed to the target-first antibody complex to form a target-first antibody-second antibody tertiary complex.
  • the complex is detected by the signal emitted by the reporter molecule.
  • reporter molecule is meant a molecule which, by its chemical nature, provides an analytically identifiable signal which allows the detection of antigen-bound antibody.
  • the most commonly used reporter molecules in this type of assay are either enzymes, fluorophores or radionuclide containing molecules (i.e. radioisotopes) and chemiluminescent molecules.
  • an enzyme is conjugated to the second antibody, generally by means of glutaraldehyde or periodate.
  • glutaraldehyde or periodate As will be readily recognized, however, a wide variety of different conjugation techniques exist, which are readily available to the skilled artisan.
  • Commonly used enzymes include horseradish peroxidase, glucose oxidase, -galactosidase and alkaline phosphatase, amongst others.
  • the substrates to be used with the specific enzymes are generally chosen for the production, upon hydrolysis by the corresponding enzyme, of a detectable color change. Examples of suitable enzymes include alkaline phosphatase and peroxidase.
  • fluorogenic substrates which yield a fluorescent product rather than the chromogenic substrates noted above.
  • the enzyme-labeled antibody is added to the first antibody-molecular marker complex, allowed to bind, and then the excess reagent is washed away. A solution containing the appropriate substrate is then added to the complex of antibody-antigen-antibody. The substrate will react with the enzyme linked to the second antibody, giving a qualitative visual signal, which may be further quantitated, usually spectrophotometrically, to give an indication of the amount of biomarker which was present in the sample.
  • fluorescent compounds such as fluorescein and rhodamine, may be chemically coupled to antibodies without altering their binding capacity.
  • the fluorochrome-labeled antibody When activated by illumination with light of a particular wavelength, the fluorochrome-labeled antibody adsorbs the light energy, inducing a state to excitability in the molecule, followed by emission of the light at a characteristic color visually detectable with a light microscope.
  • the fluorescent labeled antibody is allowed to bind to the first antibody-molecular marker complex. After washing off the unbound reagent, the remaining tertiary complex is then exposed to the light of the appropriate wavelength, the fluorescence observed indicates the presence of the molecular marker of interest.
  • Immunofluorescence and EIA techniques are both very well established in the art. However, other reporter molecules, such as radioisotope, chemiluminescent or bioluminescent molecules, may also be employed.
  • expression of a selected biomarker in a tissue or cell sample may be examined by way of functional or activity-based assays.
  • the biomarker is an enzyme
  • a sample comprising a target molecule can be obtained by methods well known in the art, and that are appropriate for the particular type and location of the disease of interest. Tissue biopsy is often used to obtain a representative piece of disease tissue. Alternatively, cells can be obtained indirectly in the form of tissues/fluids that are known or thought to contain the disease cells of interest. For instance, samples of disease lesions may be obtained by resection, bronchoscopy, fine needle aspiration, bronchial brushings, or from sputum, pleural fluid or blood. Genes or gene products can be detected from disease tissue or from other body samples such as urine, sputum or serum.
  • tissue preparation for disease cells Means for enriching a tissue preparation for disease cells are known in the art.
  • the tissue may be isolated from paraffin or cryostat sections.
  • Cells of interest may also be separated from normal cells by flow cytometry or laser capture microdissection. These, as well as other techniques for separating disease from normal cells, are well known in the art. If the disease tissue is highly contaminated with normal cells, detection of signature gene expression profile may be more difficult, although techniques for minimizing contamination and/or false positive/negative results are known, some of which are described herein below.
  • a sample may also be assessed for the presence of a biomarker (including a mutation) known to be associated with a disease cell of interest but not a corresponding normal cell, or vice versa.
  • an effective amount of the anti-CD40 antibody may be administered to the mammal, such as a human to treat a disorder, such as a B-cell lymphoma which is afflicting the mammal.
  • a disorder such as a B-cell lymphoma which is afflicting the mammal.
  • Diagnosis in mammals, such as humans, of the various pathological conditions described herein can be made by the skilled practitioner.
  • the methods described herein comprise a process of comparing a measured expression level of a marker gene and a reference level.
  • the reference level may be a measured expression level of a reference gene different from the marker gene or a measured expression level of the same marker gene in a different sample.
  • a measured expression level of a marker gene in a B cell lymphoma sample from a subject is compared to a measured expression level of a reference gene in the sample.
  • the expression level of the reference gene does not substantially change among various types of B lymphoma cells, including anti-CD40 antibody sensitive and resistant cells (e.g., genes in Table 8 or Table 9).
  • the ratio of the measured expression level of the marker gene to the measured expression level of the reference is calculated, and the ratio may be used for assessing or aiding assessment of responsiveness of the B cell lymphoma to an anti-CD antibody treatment.
  • a measured expression level of a marker gene in a B cell lymphoma sample from a subject is compared to a measured expression level of the marker gene in a reference sample.
  • the reference sample comprises B lymphoma cells that are resistant or not responsive to an anti-CD40 antibody.
  • the comparison is performed to determine the magnitude of the difference between the measured expression levels of the marker gene in the sample from the subject and in the reference sample (e.g., comparing the fold or percentage difference between the expression levels of the marker gene in the sample from the subject and the reference sample).
  • An increase or decreased expression of a marker gene in the sample from the subject as compared to the expression of the marker gene in the reference sample comprising B lymphoma cells that are resistant or not responsive to an anti-CD40 antibody suggests or indicates responsiveness of the B-cell lymphoma to treatment with an anti-CD40 antibody.
  • VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, and MEF2B are generally overexpressed in anti-CD40 antibody sensitive cells as compared to resistant cells.
  • a fold of increase in the expression level of the sample from the subject can be at least about any of 1.5 ⁇ , 1.75 ⁇ , 2 ⁇ , 3 ⁇ , 4 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ , or 10 ⁇ the expression level of the reference sample.
  • a fold of decrease in the expression level of the sample from the subject can be less than about any of 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8 of the expression level of the reference sample.
  • expression level of one or more marker genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are compared to a reference level.
  • an increased expression level of one or more of IFITM1, CD79B, IGF1R, CD44, CTSC, EPDR1, and PUS7 as compared to a reference level indicates that said subject is less likely to respond to an agonist anti-CD40 antibody treatment.
  • the reference level is a value or a range determined by expression levels of the corresponding marker gene in samples comprising B lymphoma cells from subjects having tumor volume increased after an agonist anti-CD40 antibody treatment.
  • an increased expression of one or more of CD40, RGS13, VNN2, LMO2, CD22, BTG2, and UAP1 as compared to a reference level indicates that said subject is likely to respond to the agonist anti-CD40 antibody treatment.
  • the reference level is a value or a range determined by expression levels of the corresponding marker gene in samples comprising B lymphoma cells from subjects having tumor volume decreased after an agonist anti-CD40 antibody treatment.
  • the expression level BCL6 is measured and compared to a reference level.
  • the expression level of BCL6 is used for predicting, assessing, or aiding assessment of responsiveness of the subject to an anti-CD40 antibody treatment. As shown in Example 2, BCL6 expression trends lower in those subjects with tumor increases after an agonist anti-CD40 antibody treatment. In some embodiments, an increased expression of BCL6 as compared to a reference level determined by expression level of BCL6 in samples from subjects having tumor volume decreased after an agonist anti-CD40 antibody treatment may indicate the subject is likely to respond to the agonist anti-CD40 antibody treatment.
  • the expression levels of marker genes in Table 7) are measured, and a sensitivity index calculated as the sum of signed t-scores for log 2-scale expression of genes pairs 1-8 in Table 7 is determined, wherein a sensitivity index greater than ⁇ 4 suggests or indicates the B-cell lymphoma is responsive to an anti-CD40 antibody treatment.
  • the sensitivity index is greater than ⁇ 3, greater than ⁇ 2, greater than ⁇ 1, or greater than 0.
  • the sensitivity index is between ⁇ 4 and 20. In some embodiments, the sensitivity index is between 0 and 20.
  • the expression levels of one or more of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are measured, and a sensitivity index is calculated based on the measured expression level of the marker genes.
  • a sensitivity index is calculated based on the measured expression level of the marker genes.
  • SI sensitivity index
  • expression level of at least one marker gene having a positive correlation value and at least one marker gene having a negative correlation value shown in Table 13 are measured; wherein (i) ⁇ j is the coefficient value for each marker genes measured; (ii) p is the number of marker genes measured; (iii) x i is transformed, normalized expression level for the sample from the subject for expression level of each marker measured; and (iv) ⁇ j and ⁇ j are means and standard deviations for each marker gene measured; wherein ⁇ j , ⁇ j and ⁇ j are determined from patient samples comprising B lymphoma cells from a clinical trial.
  • a value equals or greater than zero for the sensitivity index indicates that the subject is likely to respond the anti-CD40 antibody treatment, or wherein a value less than zero for the sensitivity index indicates that the subject is less likely to respond the anti-CD40 antibody treatment.
  • Example 2 described in detail how to analyze and determine parameters for reference samples and new samples.
  • the expression level of IFITM1, RGS13, CD79B, CD22, BTG2, CD44, EPDR1, and UAP1 are measured and used for the sensitivity index calculation.
  • equal number of positive correlated marker genes and negative correlated marker genes are measured and used for the sensitivity index calculation.
  • the comparison can be carried out in any convenient manner appropriate to the type of measured value and reference value for the gene markers at issue.
  • the process of comparing may be manual or it may be automatic.
  • measured expression levels are normalized values.
  • the expression level may be normalized based on the equation under Transformed, Normalized Assay Values described in Example 2.
  • replicate measurements may be taken for the expression levels of marker genes and/or reference genes.
  • replicate measurements are taking into account for the measured values.
  • the replicate measurements may be taken into account by using either the mean or median of the measured values as the “measured value”. Statistical analysis known in the art may be used to verify the significance of the difference between the two values compared.
  • the marker genes identified in the invention may be used for predicting, assessing, or aiding assessment of responsiveness of B-cell lymphoma to treatment with one or more anti-CD40 antibodies.
  • the anti-CD40 antibodies may be one or more agonist antibodies (i.e., bind and stimulate CD40).
  • Stimulatory antibodies can be of different types, such as: (1) those that deliver a stimulatory signal through CD40 but do not increase the interaction between CD40 and CD40L (e.g., antibody G28-5 and antibodies derived from G28-5 described in U.S. Pat. No.
  • CD40 and CD40L e.g., antibodies HuCD40-M2 and HuCD40-M3 and humanized antibodies described in U.S. Pat. No. 5,674,492; and (2) those that deliver a stimulatory signal through CD40 and can increase the interaction between CD40 and CD40L, e.g., S2C6 (Francisco et al., 2000 , Cancer Res. 60:3225-31) and antibodies derived from S2C6. Agonists antibodies are also described in U.S. Pat. No. 7,288,251.
  • the anti-CD40 antibodies may be one or more antagonist antibodies (i.e., bind CD40 and inhibit activities induced by CD40L). Examples of antagonist anti-CD40 antibodies include human antibody CHIR-12.12 described in U.S. Pub. No. 2007/0110754, and anti-CD40 antibodies described in WO 97/31025.
  • the methods of the invention may further comprise administering an effective amount of an anti-CD40 antibody to a subject having a B-cell lymphoma after the subject has been identified as a candidate for treatment based on the assays/methods described herein.
  • One or more anti-CD40 antibodies may be administered.
  • the anti-CD40 antibody is administered in conjunction with one or more of the following therapeutic agents: rituximab, gemzar, and ICE.
  • an anti-CD40 antibody can be administered to the patient in conjunction with rituximab therapy; with rituximal plus gemzar; with rituximal plus ICE (ifosfamide, carboplatin, etoposide) (R-ICE); or with rituximab plus chemotherapy.
  • rituximab therapy with rituximal plus gemzar; with rituximal plus ICE (ifosfamide, carboplatin, etoposide) (R-ICE); or with rituximab plus chemotherapy.
  • administration “in conjunction” includes simultaneous administration and/or administration at different times.
  • Administration in conjunction also encompasses administration as a co-formulation (i.e., different drugs are present in the same composition) or administration as separate compositions, administration at different dosing frequencies or intervals, and administration using the same route or different routes.
  • the anti-CD40 antibodies or functional fragments can be used for the treatment of patients with NHL that are nonresponsive or have an inadequate response to treatment with any one of the following drugs: rituximab (Genentech); ocrelizumab (Genentech, Inc.); ibritumomab tiuxetan (ZevalinTM, Biogen Idec); tositumomab (BexxarTM, GlaxoSmithKline); HuMAX-CD20TM (GenMab); IMMU-106 (which is a humanized anti-CD20 a.k.a.
  • hA20 or 90Y-hLL2, Immunomedics AME-133 (Applied Molecular Evolution/Eli Lilly); gentuzumab ozogamicin (MylotargTM, a humanized anti-CD33 antibody, Wyeth/PDL); alemtuzumab (CampathTM, an anti-CD52 antibody, Schering Plough/Genzyme); epratuzumab (IMMU-103TM, a humanized anti-CD22 antibody, Immunomedics), or have relapsed after treatment with these drugs.
  • MylotargTM a humanized anti-CD33 antibody
  • Wyeth/PDL Wyeth/PDL
  • alemtuzumab CampathTM, an anti-CD52 antibody, Schering Plough/Genzyme
  • epratuzumab IMMU-103TM, a humanized anti-CD22 antibody, Immunomedics
  • lymphomas and CLL their diagnoses, treatment and standard medical procedures for measuring treatment efficacy.
  • Anti-CD40 antibodies for use in the treatment include chimeric, humanized and human antibodies. Any agonist or antagonist antibodies described herein or known in the art may be used in the treatment. For example, humanized anti-CD40 antibodies described in WO 2006/128103 may be used for the anti-CD40 antibody treatment, and these antibodies and their amino acid sequences are incorporated herein by reference.
  • the anti-CD40 antibody for used in the treatment described herein binds to CD40 (such as human CD40) expressed on B lymphoma cells and induces apoptosis of the B lymphoma cells.
  • the anti-CD40 antibody may also have the characteristics of killing B lymphoma cells in vivo via immune effector functions, such as ADCC, CDC, and/or ADCP.
  • the anti-CD40 antibody binds to CD40 with a K d value of no higher than about 1 ⁇ 10 ⁇ 8 or no higher than 1 ⁇ 10 ⁇ 9 . In some embodiments, the anti-CD40 antibody binds to CD40 and stimulates CD40 (i.e., an agonist antibody). In some embodiments, the anti-CD40 antibody increases the binding of CD40 ligand to CD40, for example, by at least 45%, by at least 50%, by at least 60%, or by at least 75%. A method of determining increases in binding of CD40 ligand to CD40 are disclosed in U.S. Pat. No. 6,838,261 (the disclosure of which is incorporated by reference herein).
  • the anti-CD40 is a humanized antibody derived from murine monoclonal antibody S2C6 described in WO 00/75348 (including antibodies provided in Tables 3 and 4 of WO 00/75348).
  • the anti-CD40 antibody comprises the heavy chain amino acid sequence shown in SEQ ID NO:1 and the light chain amino acid sequence shown in SEQ ID NO:2, for example anti-CD40 Ab.1.
  • kits or articles of manufacture are also provided by the invention.
  • Such kits may comprise at least one reagent specific for detecting expression level of a marker gene described herein, and may further include instructions for carrying out a method described herein.
  • the invention provides compositions and kits comprising primers and primer pairs, which allow the specific amplification of the polynucleotides of the invention or of any specific parts thereof, and probes that selectively or specifically hybridize to nucleic acid molecules of the invention or to any part thereof.
  • Probes may be labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme.
  • Such probes and primers can be used to detect the presence of polynucleotides, such as the polynucleotides corresponding to genes listed in Table 1-4, 6, 7 and 13, in a sample and as a means for detecting a cell expressing proteins encoded by the polynucleotides corresponding to genes listed in Table 1-4, 6, 7 and 13.
  • polynucleotides such as the polynucleotides corresponding to genes listed in Table 1-4, 6, 7 and 13
  • a great many different primers and probes may be prepared based on the sequences provided in herein and used effectively to amplify, clone and/or determine the presence and/or levels of mRNAs.
  • kits comprise reagents for detecting expression levels of at least two, at least three, at least five, at least ten, at least fifteen, at least twenty marker genes. Kits may also comprise reference samples that are useful as generating reference values.
  • the marker genes include, but are not limited to VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, MEF2B, LRRC8A, CD40, IFITM1, SMN1, PRRCA, EPDR1, PRPSAP2, IGF1R, BTG2, LMO2, YIPF3, CD79B, CD44, CTSC, UAP1, and PUS7.
  • the reagents for detecting mRNA expression level of a marker gene may comprise at least one pair of primers specific for amplifying the mRNA products of one marker gene.
  • the pair of primers may target the 3′ end of the mRNA sequence (e.g., targeting mRNA at the 3′ UTR which is usually shared in common with all transcript variants).
  • the kits may further comprise a surface or substrate (such as a microarray) for capture probes for detecting of amplified nucleic acids.
  • kits comprises at least one pair of primers and a probe specific for detecting one marker gene expression level using qRT-PCR.
  • sets of primers and probes that can be used in qRT-PCR are shown in Table 10.
  • primer and probe sets shown in SEQ ID NOS:27, 28 and 29, SEQ ID NOS:60, 61, and 62, and SEQ ID NOS:93, 94, and 95 may be used.
  • primer and probe sets shown in SEQ ID NOS:24, 25, and 26, SEQ ID NOS:57, 58, and 59, SEQ ID NOS:90, 91 and 92 may be used.
  • primer and probe sets shown in SEQ ID NOS:114, 115, and 116, and SEQ ID NOS:126, 127, and 128 may be used.
  • VNN2 primer and probe sets shown in SEQ ID NOS:30, 31, and 32, SEQ ID NOS:63, 64, and 65, and SEQ ID NOS:96, 97, and 98.
  • LMO2 primer and probe sets shown in SEQ ID NOS:12, 13, and 14, SEQ ID NOS:45, 46, and 47, and SEQ ID NOS:78, 79, and 80.
  • primer and probe sets shown in SEQ ID NOS:6, 7, and 8, SEQ ID NOS:39, 40, and 41, and SEQ ID NOS:72, 73, and 74 For detecting CD44, primer and probe sets shown in SEQ ID NOS:174, 175, and 176, SEQ ID NOS:180, 181, and 182, and SEQ ID NOS:186, 187, and 188.
  • the reagents for detecting protein expression level of a marker gene may comprise an antibody that specifically binds to the protein encoded by the marker gene.
  • kits may further comprise a carrier means being compartmentalized to receive in close confinement one or more container means such as vials, tubes, and the like, each of the container means comprising one of the separate elements to be used in the method.
  • container means such as vials, tubes, and the like
  • each of the container means comprising one of the separate elements to be used in the method.
  • one of the container means may comprise a probe that is or can be detectably labeled.
  • probe may be an antibody or polynucleotide specific for a marker gene.
  • the kit may also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence and/or a container comprising a reporter-means, such as a biotin-binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, florescent, or radioisotope label.
  • a reporter-means such as a biotin-binding protein, such as avidin or streptavidin
  • the kit of the invention will typically comprise the container described above and one or more other containers comprising materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a label may be present on the container to indicate that the composition is used for a specific therapy or non-therapeutic application, and may also indicate directions for either in vivo or in vitro use, such as those described above.
  • the kit can further comprise a set of instructions and materials for preparing a tissue or cell sample and preparing nucleic acid (such as mRNA) from the sample.
  • nucleic acid such as mRNA
  • an array of the invention comprises individual or collections of nucleic acid molecules useful for detecting mutations of the invention.
  • an array of the invention may comprises a series of discretely placed individual nucleic acid oligonucleotides or sets of nucleic acid oligonucleotide combinations that are hybridizable to a sample comprising target nucleic acids, whereby such hybridization is indicative of presence or absence of a mutation of the invention.
  • nucleic acids attaching nucleic acids to a solid substrate such as a glass slide.
  • One method is to incorporate modified bases or analogs that contain a moiety that is capable of attachment to a solid substrate, such as an amine group, a derivative of an amine group or another group with a positive charge, into nucleic acid molecules that are synthesized.
  • the synthesized product is then contacted with a solid substrate, such as a glass slide, which is coated with an aldehyde or another reactive group which will form a covalent link with the reactive group that is on the amplified product and become covalently attached to the glass slide.
  • Other methods such as those using amino propryl silican surface chemistry are also known in the art, as disclosed at world wide web at cmt.corning.com and cmgm.stanford.edu/pbrown1.
  • Attachment of groups to oligonucleotides which could be later converted to reactive groups is also possible using methods known in the art. Any attachment to nucleotides of oligonucleotides will become part of oligonucleotide, which could then be attached to the solid surface of the microarray. Amplified nucleic acids can be further modified, such as through cleavage into fragments or by attachment of detectable labels, prior to or following attachment to the solid substrate, as required and/or permitted by the techniques used.
  • NHL Cells were seeded in 384 well plates at 1500-5000 cells/well in 50 ul RPMI 1640 supplemented with 2% FBS and treated with serial concentrations of crosslinked anti-CD40 Ab.1 or control antibody (anti-gD 5B6).
  • anti-CD40 Ab.1 or anti-gD was incubated with F(ab′)2 fragments of a goat anti human IgG Fc ⁇ fragment-specific antibody (Jackson ImmunoResearch, West Grove, Pa.) in a 1:4 ratio in medium for 30 minutes at room temperature before adding to cells. After 96 hours of incubation, cell viability was evaluated using CellTiter-Glo Luminescent Cell Viability Assay (Promega, Madison, Wis.) according to the manufacturer's instructions. Each data point was performed in quadruplicate.
  • Sensitivity to anti-CD40 Ab.1 was binned into three categories: Sensitive, Intermediate, and Resistant based on IC25 and IC50 values.
  • anti-CD40 Ab.1 is a humanized IgG1 mAb against CD40. It is produced in and secreted by a genetically engineered Chinese Hamster Ovary (CHO) cell line.
  • the anti-CD40 Ab.1 used in the examples and referred to as anti-CD40 Ab.1 has the following amino acid sequence:
  • DIQMTQSPSS LSASVGDRVT ITCRSSQSLV HSNGNTFLHW YQQKPGKAPK 50 LLIYTVSNRF SGVPSRFSGS GSGTDFTLTI SSLQPEDFAT YFCSQTTHVP 100 WTFGQGTKVE IKRTVAAPSV FIFPPSDEQL KSGTASVVCL LNNFYPREAK 150 VQWKVDNALQ SGNSQESVTE QDSKDSTYSL SSTLTLSKAD YEKHKVYACE 200 VTHQGLSSPV TKSFNRGEC 219
  • Probes were mapped to each gene and a 1:1 probe to gene mapping was selected for downstream analysis using the probe most strongly associated with the measure of sensitivity.
  • quantitative stepwise linear modeling was combined with qualitative analysis of target pathways to identify a parsimonious set of genes to inclusion in the assay. Further details and results are provided in the Results (Table 7).
  • Gene set enrichment analysis was determined by utilizing the GSEA module within Gene Pattern (world wide web at genepattern.org).
  • the enrichment score awards pre-specified classes of genes when their members are significantly differentially expressed in a concordant manner across phenotypes.
  • the normalized enrichment score is calculated by taking the enrichment score and adjusting for the number of genes within a gene set.
  • the nominal p-value is determined by permutating the sensitive and resistant labels and recomputing the normalized enrichment score to give a null distribution.
  • Each Target Gene is shown with its corresponding inversely correlated (anti-correlated) Pair Gene (Table 7), in order of the step at which the Target Gene was chosen for inclusion in the Index.
  • the first 3 Main Genes (VNN2, RGS13, CD22 in Table 7) were selected from Tables 2-4 (Step 1) to model the dominant component of differential over-expression in Sensitive and Intermediate cell lines.
  • the expression of these 3 genes is highly correlated, with correlation coefficients of +0.77 or higher. Due to their similarity, a single pair gene EPDR1 was selected from Tables 2-4 to measure contrasting overexpression in Resistant cell lines. Including such anti-correlated Pair Genes in the assay provides auto-normalization in that both Sensitivity and Resistance are associated with high expression of one arm of the pair.
  • the assay does not depend upon low overall mRNA assay levels to define any class, but rather describes each by a pattern of relative expression of the Main Genes to their anticorrelated Pairs (i.e. a sum of signed t-scores on the log 2 scale, with signs corresponding to the Fold Change Estimate).
  • additional pairs of genes were chosen based upon mechanism of action from a new list of those with significant associations to IC25 after adjustment for the cumulative sum of signed t-scores for genes identified in previous Steps. This stepwise procedure requires each new pair of genes to add additional predictive power to the Sensitivity Index. After Step 5, no more gene pairs were needed for IC25 prediction.
  • Step 6 a single additional pair was added for its ability to predict cell viability at maximum inhibition after adjusting for the cumulative Index based upon the previous 7 pairs of genes.
  • BCL6 was added as a singleton without a corresponding pair based upon a mechanism of action rationale: it is not currently incorporated in the final Sensitivity Index, which is given by the sum of signed t-scores for log 2-scale expression of Gene Pairs 1-8. It may be incorporated explicitly into the index based upon clinical experience. For classification into Sensitive or Intermediate versus Resistant groups, a preliminary cutoff was chosen for the Sensitivity Index so as to maximize the overall correct classification rate. Alternate classification rules based upon the selected probes may be optimized later for clinical application.
  • Table 1 provides anti-CD40 Ab.1 IC25 sensitivity data across NHL cell lines in vitro. Specific lymphoma subtypes of each cell line, IC25 values and classifier data are given for each cell line.
  • DLBCL Diffuse Large B-cell Lymphoma
  • FL Follicular Lymphoma
  • MCL Mantle Cell Lymphoma
  • ALCL Anaplastic Large Cell Lymphoma
  • RNA was prepared from the cell lines at the log stage of cell division and subjected to gene expression profiling using the Affymetrix HGU133P2 microarray. Differentially expressed genes between Sensitive and Resistant cell lines were determined by a moderated t-test and significance was determined using an adjusted P-value cutoff of ⁇ 0.05 (Table 2). In Table 2, gene list filtered to an adjusted p-value ⁇ 0.05 (5% FDR) resulting in 110 unique genes. Probe ID, gene symbol and description are indicated.
  • SEMA4C 46665_at sema domain NA NA 0.658 immunoglobulin domain (Ig), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 4C ZNF322A 219376_at zinc finger protein 322A NA NA 0.691
  • the genes that are highly expressed in Table 4 may be co-regulated genes that may not be related to the biology of anti-CD40 activity. Therefore to comprehend the biological function of the genes that are differentially expressed between sensitive and resistant cells, we carried out Gene Set Enrichment Analysis (GSEA).
  • GSEA Gene Set Enrichment Analysis
  • Table 5 provides gene set enrichment analysis of anti-CD40 Ab.1 Sensitive vs.
  • Resistant NHL cell lines Enriched gene sets, number of genes per gene set, normalized enrichment score (NES), and nominal p-value (NOM p-val) are displayed. The higher the NES and the lower the NOM p-val, the more likely the findings are significant.
  • NES normalized enrichment score
  • NOM p-val nominal p-value
  • Ranked genes are derived from the moderated t-test (Table 2). 70 genes in total were part of this gene set with the top 11 being displayed in this table. Genes shown in table 6 were overexpressed in anti-CD40 Ab.1 sensitive cell lines. The partial overlap of genes with the BCR and CD40L genes is expected since the two signal transduction pathways converge at the axis of NF- ⁇ B transcription and both pathways can synergize to activate B-cells. We next ascertained if any of the CD40L-induced genes are capable of discriminating between sensitive and resistant NHL cell lines to anti-CD40 Ab.1. Of the CD40L genes within the differentially expressed gene list on Tables 2 and 3, VNN2 gave the most accurate discrimination for sensitive and resistant cell lines ( FIG. 2 ).
  • CD40L plays a critical role in activating B-cells and results in the expansion and proliferation of B-cells as well as Ig class switching and the CD40L signaling pathway is also active within pre- and post-GCB-cells including na ⁇ ve and memory B-cells. Therefore, it is striking to note that NHL cells that are displaying sensitivity to anti-CD40 Ab.1 are similar to GCB-cells in origin by gene expression profiling and have CD40L downregulated genes highly expressed, in contrast to resistant cells, indicative of a relationship between GCB and CD40 pathway activation status determining sensitivity to anti-CD40 Ab. 1.
  • xenograft models are used to explore in therapy (such as combination therapy).
  • Real time quantitative RT-PCR (qRT-PCR) is used for measuring gene expression levels.
  • immunohistochemistry (IHC) assays are developed for a small group of markers selected (e.g., VNN2 and RGS13). Selected marker genes are further tested in clinical trial samples.
  • qRT-PCR and IHC are performed to measure expression levels of selected marker genes in clinical trial samples. Expression levels in samples from patients having relapsed diffuse large B-cell lymphoma that are responsive to the anti-CD40 treatment are compared the expression levels in samples from patients that are not responsive to the treatment.
  • Tumor tissues were taken from patients before they received treatment with anti-CD40 Ab.1. For example, samples were taken as part of routine lymphoma diagnosis.
  • Multi-institutional, multi-dose phase I study was conducted to test the safety, pharmacokinetic properties, immunogenicity, and antitumor activity of intravenous anti-CD40 Ab.1 in patients with relapsed NHL.
  • Patients with multiple histologic subtypes of NHL were enrolled on this study, including diffuse large B-cell (DLBCL; 14), follicular (FCL; 9), mantle cell (MCL; 9), marginal zone (MZL; 2) and small lymphocytic (SLL; 1).
  • Patients were treated with a dose-loading schedule: 1 mg/kg of anti-CD40 Ab.1 on day 1 and day 4 and subsequent intra-patient dose-escalation during weeks 2-5 to a maximum dose of 3, 4, 6, or 8 mg/kg over four cohorts.
  • a rapid dose-loading schedule was tested in one cohort (40% increase in total anti-CD40 Ab.1 administered during cycle 1). Responding patients or those with stable disease were eligible for a second cycle, consisting of four consecutive weekly infusions at the cohort-specific maximum dose of anti-CD40 Ab.1. Eight patients with DLBCL completed cycle 1 and received a maximum dose of at least 3 mg/kg anti-CD40 Ab.1 with an objective response rate of 37.5% (i.e. 1 CR and 2 PR) and 2 SD. Additional objective responses were seen in one patient with MCL (CR) and one patient with MZL (PR). The median duration of response for these 5 patients has not yet been reached (range 8-37 weeks). Tumor tissues were taken from patients before they received treatment with anti-Cd40 Ab.1. For example, samples were taken as part of routine lymphoma diagnosis.
  • Formalin Fixed Paraffin Embedded (FFPE) archival tumor tissue from the Phase I and Phase II clinical trials described above was obtained from the clinical investigation sites with appropriate IRB approval and patient consent.
  • FFPE Formalin Fixed Paraffin Embedded
  • 4-6 micron sections derived from the tumor tissue were mounted on glass slides and one slide for each case was subject to H&E staining using standard pathology laboratory protocol.
  • a board certified Pathologist marked the H&E slide for tumor content and was used as a guide to macrodissect the remaining tumor-containing region for RNA extraction using the Ambion RecoverAllTM Total Nucleic Acid Isolation Kit for FFPE Tissues (Cat. No. AM1975; Applied Biosystems/Ambion, Austin, Tex.).
  • RNA per sample was reverse transcribed in a total reaction volume of 20 uL using Applied Biosystems' High Capacity Reverse Transcription cDNA Synthesis kit (Cat. No. 4368814; Applied Biosystems, Foster City, Calif.). Manufacturer's recommendations were followed with the exception of a shortened 60 min RT reaction at 37 degrees. 5 ng total RNA equivalent cDNA (assuming 100% cDNA synthesis efficiency) product was mixed with Applied Biosystems' 2 ⁇ Universal Master Mix (no UNG) in a volume of 15 uL for each PCR assay well. All amplifications were performed in triplicate in 384-well plates using a 2-step (95 degrees 15 sec, 60 degrees 1 min) PCR amplification procedure. Reactions were carried out to 40 cycles on a validated ABI 7900 real-time PCR system. Sequences of the primers and probes used are shown in Table 10.
  • Table 11 provides a sample accounting of assayed specimens and clinical samples from Clinical Trial 001.
  • Twenty nine archival FFPE tumor specimens from 24 patients with DLBCL were submitted for qRT-PCR processing. Three patients had multiple specimens and all 24 patients had usable qRT-PCR results for at least one specimen. Of these 24, 21 had tumor sum of the product of diameters (SPD) measurements reported both at baseline and at least one post-baseline visit.
  • SPD product of diameters
  • Narrower ranges will become available as the sample size increases. Since no model-building or checking was required to produce these results, they comprise a robust trend, which confirms that these qRT-PCR probe measurements are associated, overall, with reduction in tumor SPD in patients treated with anti-CD40 Ab. 1.
  • the multivariate sensitivity index is a weighted average of the probes in Tables 12 and 13. Since weights in cell lines were not expected to reflect optimal weights in patient tumor specimens, the weights in cell lines were restricted to 1 and ⁇ 1, corresponding to the signed, equal-weighted average, where the signs matched the association between each probe and resistance to anti-CD40 Ab.1 by IC25 in the cell lines. For clinical populations, new weights are required. As a preliminary analysis based upon 21 samples only, we chose to use a penalized, multivariate regression procedure to select and estimate weights for the best 8 of the 14 probes. Those weights (coefficient) are shown in Table 14, and the association between the resulting Sensitivity Index and SPD change from baseline is depicted in FIG. 7 .
  • Raw qRT-PCR results were successfully generated for 10 patients with archival specimens. For those 10 patients, diagnosis, treatment group, multivariate sensitivity index, clinical response and SPD change from baseline are shown in Table 16.
  • the multivariate sensitivity index weights were taken from the 21 Clinical Trial 001 patients (Table 14), so that these patients constitute a very small validation set. 2 of 4 patients with Sensitivity Index ⁇ 0 exhibited some tumor shrinkage after anti-CD40 Ab.1 exposure and 4 of 6 patients with Sensitivity Index ⁇ 0 exhibited either tumor increase or a best response of PD (SPD was unavailable for 2 patients, but a best clinical response outcome was available for this patient).

Abstract

The invention provides methods and kits useful for predicting or assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies.

Description

RELATED APPLICATIONS
This application is a National Phase application under 35 U.S.C. §371 of International Application No. PCT/US2008/082920, filed on Nov. 7, 2008, which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
The present invention relates generally to the fields of predicting, assessing, aiding assessment of responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies.
BACKGROUND
CD40 is a type I transmembrane protein of the tumor necrosis receptor superfamily. CD40 is an important molecule involved in B-cell proliferation and differentiation, immunoglobulin isotype switching, and cell viability. Receptor signaling is initiated by the binding of CD40 to the CD40 ligand (CD40L or CD154), which is primarily expressed on activated CD4+ T cells.
On normal cells, CD40 is expressed on cells with high proliferative potential, including hematopoietic progenitors, epithelial and endothelial cells, and all antigen-presenting cells (dendritic cells, activated B lymphocytes, and activated monocytes). CD40 is highly expressed on several types of B-cell hematologic malignancies including multiple myeloma, non-Hodgkin's lymphoma (NHL), and chronic lymphocytic leukemia (CLL). The high prevalence of CD40 expression on B-cell malignancies makes it an attractive potential tumor target for antibody-based cancer therapy. CD40 is also expressed on a majority of bladder cancers and a significant percentage of other solid tumors, including head and neck cancers, renal cell carcinomas, ovarian and lung cancer.
Anti-CD40 antibodies and their uses for treating B cell hematologic malignancies have been described. See, e.g., U.S. Pat. Nos. 6,946,129; 6,843,989; 6,838,261; WO 2000/075348; US-2002-0197256; WO 2006/128103; and WO 2007/075326. It has been shown that a humanized anti-CD40 antibody induces growth inhibition and apoptosis of CD40-positive cells in a subset of hematologic tumor cell lines through direct signal transduction. WO 2006/128103; WO 2007/075326. Furthermore, the humanized anti-CD40 antibody kills tumor cells via immune effector functions, including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). In vivo, using xenograft models of multiple myeloma (MM) and non-Hodgkin's lymphoma (NHL), the anti-CD40 antibody suppresses tumor growth and improves survival in severe combined immunodeficient (SCID) mice. Comparison of the anti-CD40 antibody to rituximab (Genentech, Inc.) in several models revealed anti-tumor activity of the anti-CD40 antibody was at least as effective as rituximab.
Seattle Genetics initiated Phase I clinical trials in 2004 with the humanized anti-CD40 antibody in a single agent multi-dose trial in patients with relapsed and refractory multiple myeloma (MM). Subsequently, Phase I trials were initiated in patients with relapsed non-Hodgkin's lymphoma (NHL) and chronic lymphocytic lymphoma (CLL). The results from these Phase I trials showed evidence for anti-tumor activity in myeloma patients with stable disease and decreased M-protein, NHL patients with partial and complete responses, and CLL patients with stable disease. A phase II trial of the anti-CD40 antibody in relapsed diffuse large B cell lymphoma (DLBCL) was initiated in December 2006.
Although it has been shown anti-CD40 antibodies can induce growth inhibition and apoptosis of CD40-positive cells and may have anti-tumor activity in various types of B cell lymphoma patients, not all B lymphoma cells are sensitive to anti-CD40 antibody mediated cell death. There remains a need to identify one or more predictive markers for the responsiveness of B-cell lymphoma patients to anti-CD40 antibody therapy.
All references cited herein, including patent applications and publications, are incorporated by reference in their entirety.
SUMMARY OF THE INVENTION
The invention provides methods and compositions for predicting, assessing or aiding assessment of responsiveness of a subject having a type of B-cell lymphoma to treatment with an anti-CD40 antibody.
In one aspect, the invention provides methods for assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to treatment with an anti-CD40 antibody, comprising comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7 and 13 in a B-cell lymphoma sample from the subject to a reference level.
In another aspect, the invention provides methods for predicting responsiveness or monitoring treatment/responsiveness to an anti-CD40 antibody treatment in a subject having a B-cell lymphoma, comprising comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7 and 13 in a B-cell lymphoma sample from the subject to a reference level.
In another aspect, the invention provides methods for predicting, assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to an anti-CD40 antibody treatment, comprising the steps of: (a) measuring expression level of one or more marker genes in a sample comprising B lymphoma cells obtained from said subject, wherein said one or more marker genes are selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7; (b) predicting whether the subject is likely to respond to the anti-CD40 antibody treatment based on the measured expression level of said one or more marker genes from step (a). In some embodiments, expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen maker genes from the group are measured and used for the prediction, assessment, or aiding assessment. In some embodiments, the prediction, assessment, or aiding assessment is determined by comparing the measured expression level of one or more marker genes to a reference level. In some embodiments, a reference level is a value or a range determined based on the measured expression level of the corresponding marker gene in samples comprising the B lymphoma cells from subjects having tumor volume increased or decreased after the anti-CD40 antibody treatment.
In another aspect, the invention provides methods preparing a personalized genomics profile for a subject having B-cell lymphoma comprising the steps of: (a) determining expression level of one or more marker genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, PUS7, and BCL6 in a sample comprising B lymphoma cells obtained from the subject; and (b) generating a report summarizing the expression level of one or more marker genes obtained in step (a). In some embodiments, expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or fifteen maker genes from the group are measured and used for the generating the report for the personalized genomics profile. In some embodiments, the report includes a recommendation for an anti-CD40 antibody treatment for the subject. In some embodiments, the recommendation is determined by comparing the measured expression level of the marker genes to a reference level. In some embodiments, a reference level is a value or a range determined based on the measured expression level of the corresponding marker gene in samples comprising the B lymphoma cells from subjects having tumor volume increased or decreased after the anti-CD40 antibody treatment.
In another aspect, the invention provides methods for predicting, assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to an anti-CD40 antibody treatment, comprising the steps of: (a) measuring expression level at least two marker genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 in a sample comprising B lymphoma cells from the subject; (b) calculating sensitivity index value (SI) based on the measured expression level of the marker genes in step (a) by the following equation:
SI = j = 1 p β j x j - μ ^ j σ ^ j 2
wherein expression level of at least one marker gene having a positive correlation value and at least one marker gene having a negative correlation value shown in Table 13 are measured;
wherein (i) βj is the coefficient value for each marker genes measured; (ii) p is the number of marker genes measured; (iii) xi is transformed, normalized expression level for the sample from the subject for expression level of each marker measured; and (iv) μj and σj are means and standard deviations for each marker gene measured; wherein βj, μj and σj are determined from patient samples comprising the B lymphoma cells. In some embodiments, a value equals or greater than zero for the sensitivity index indicates that the subject is likely to respond the anti-CD40 antibody treatment, or wherein a value less than zero for the sensitivity index indicates that the subject is less likely to respond the anti-CD40 antibody treatment. In some embodiments, the expression level of at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen marker genes are measured and used for the sensitivity index calculation. In some embodiments, the expression level of IFITM1, RGS13, CD79B, CD22, BTG2, CD44, EPDR1, and UAP1 are measured and used for the sensitivity index calculation.
In another aspect, the invention provides methods for treating a subject having a B-cell lymphoma, comprising administering an effective amount of the an anti-CD40 antibody to the subject, wherein the responsiveness of the B-cell lymphoma in the subject has been assessed by the methods described herein. In another aspect, the invention provides methods for treating a subject having a B-cell lymphoma, comprising a) selecting a subject for an anti-CD40 antibody treatment by comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7 and 13 in a B-cell lymphoma sample from the subject to a reference level to assess if the B-cell lymphoma in the subject is suitable for the anti-CD40 antibody treatment; and administering an effective amount of the anti-CD40 antibody to the subject.
In some embodiments, the reference level is a measured expression level of one or more reference genes in Table 8 or Table 9 in the B-cell lymphoma sample from the subject.
In some embodiments, the reference level is a measured expression level of the marker gene in a different B-cell lymphoma sample. In some embodiments, the different B cell lymphoma sample comprises B lymphoma cells that are resistant to an anti-CD40 antibody induced cell death.
In some embodiments, the measured expression level of the marker gene and/or the reference level are normalized.
In some embodiments, measured expression levels of at least two, at least five, at least ten, at least fifteen, or at least twenty genes in any of Tables 2-4, 6, 7 and 13 in the B-cell lymphoma sample from the subject are compared to one or more reference levels.
In some embodiments, the expression level is measured by detecting mRNA expression (e.g., real time quantitative reverse transcription PCR (qRT-PCR)) and/or by detecting protein expression (e.g., immunohistochemistry (IHC)).
In some embodiments, the marker genes measured comprise one or more CD40 ligand downregulated genes (e.g., VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, and POU2AF1). In some embodiments, the marker genes measured comprise one or more genes in the B-cell receptor signaling pathway (e.g., CD22, RGS13, and MEF2B).
In some embodiments, expression levels of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen genes selected from the group consisting of VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, and MEF2B in the B-cell lymphoma sample from the subject are compared to one or more reference levels.
In some embodiments, expression levels of one or more gene pairs selected from the group consisting of VNN2 and EPDR1, RGS13 and EPDR1, CD22 and EPDR1, LRRC8A and PRPSAP2, CD40 and IGF1R, IFITM1 and BTG2, SMN1 and LMO2, PRKCA and YIPF3 in a the B-cell lymphoma sample are compared. In some embodiments, expression levels are compared between one or more gene pairs VNN2 and EPDR1, RGS13 and EPDR1, CD22 and EPDR1, LRRC8A and PRPSAP2, CD40 and IGF1R, IFITM1 and BTG2, SMN1 and LMO2, PRKCA and YIPF3 in the B-cell lymphoma sample, and sensitivity index calculated as the sum of signed t-scores for log 2-scale expression of the gene pairs is used to assess responsiveness of the B-cell lymphoma to an anti-CD40 antibody treatment.
In some embodiments, the B-cell lymphoma is non-Hodgkin's lymphoma (NHL), including, but is not limited to, follicular lymphoma, relapsed follicular lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, lymphoplasmacytic lymphoma, mycosis fungoides/Sezary syndrome, splenic marginal zone lymphoma, and diffuse large B-cell lymphoma. In some embodiments, the B-cell lymphoma is selected from the group consisting of indolent lymphoma, aggressive lymphoma, and highly aggressive lymphoma.
In a further aspect, the invention provides kits comprising reagents for measuring expression levels of at least one marker gene in any of Tables 2-4, 6, 7 and 13. In some embodiments, the kits comprise at least a pair of primers for amplifying by PCR at least one marker gene in any of Tables 2-4, 6, 7 and 13. For example, forward and reverse primers shown in Table 10 may be used. The kits may further comprise a pair of primers for amplifying a reference gene in Table 8. The kits may further comprise a surface having attached thereof probes for detecting the amplified gene products, such as a microarray and the invention contemplates and includes such surfaces. In some embodiments, the kits comprise at least a pair of primers and a probe for detecting expression level of one marker gene in any of Tables 2-4, 6, 7 and 13 by qRT-PCR. The kits may further comprise a pair of primers and a probe for detecting expression level of a reference gene in Table 8 by qRT-PCR. For example, primer and probe sets shown in Table 10 may be used for detection expression level of genes by qRT-PCR. In some embodiments, the kits comprise one or more antibodies that specifically recognize one or more proteins encoded by the marker gene. The kits may further comprise other reagents and/or instructions for carrying out any of the methods described herein.
It is to be understood that one, some, or all of the properties of the various embodiments described herein may be combined to form other embodiments of the present invention. These and other aspects of the invention will become apparent to one of skill in the art.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1. Enrichment plot of genes within BASSO_GERMINAL_CENTER_CD40_DN gene set. The upper plot represents the enrichment score distribution across the ranked genes from the moderated t-test (Table 2). The lower plot displays the distribution of the enrichment with respect to a ranked list metric known as signal2noice. Overall, these plots clearly show that the gene set is strongly enriched within anti-CD40 Ab.1 sensitive cells.
FIG. 2. VNN2, a CD40L-downregulated gene, is overexpressed in sensitive NHL cells to anti-CD40 Ab.1 and discriminates between the two classes of sensitive and resistant. The bar graph represents the mRNA expression level and the line graph represents the IC25 values.
FIG. 3A-3C. RGS13, CD22, and MEF2B germinal center B markers, are overexpressed in sensitive and intermediate NHL cells to anti-CD40 Ab.1 and can discriminate with reasonable accuracy between the two classes of sensitive and resistant. The bar graph represents the mRNA expression level and the line graph represents the IC25 values.
FIG. 4. Anti-CD40Ab.1 Sensitivity Index Scoring Across NHL Cell Lines. Stepwise Linear Modeling and gene-pair scoring was applied to each cell line based on mRNA expression data. The primary y-axis displays the anti-CD40 Ab.1 Sensitivity Index and the secondary y-axis displays the anti-CD40 Ab.1 IC25 values plotted against the NHL cell lines on the x-axis. A high anti-CD40 Ab.1 Sensitivity Index (>−4) represents an increased probability of a cell line being sensitive.
FIG. 5. Correlation of CD40 signature genes with anti-CD40.Ab.1 sensitivity.
FIGS. 6-1 to 6-35. Gene bank sequences for genes listed in Table 7 and Table 10. Nucleic acid sequences encoding mRNA of VNN2 (FIG. 6-1: SEQ ID NO:258), RGS13 (FIG. 6-2: SEQ ID NO:259), CD22 (FIGS. 6-3 and 6-4: SEQ ID NO:260), LRRC8A (FIG. 6-5: SEQ ID NO:261), CD40 (FIG. 6-6: SEQ ID NO:262), IFITM1 (FIG. 6-7: SEQ ID NO:263), PRKCA (FIGS. 6-8 to 6-10: SEQ ID NO:264), BCL6 (FIGS. 6-11 and 6-12: SEQ ID NO:265), EPDR1 (FIG. 6-13: SEQ ID NO:266), PRPSAP2 (FIG. 6-14: SEQ ID NO:267), IGF1R (FIGS. 6-15 to 6-18: SEQ ID NO:268), BTG2 (FIGS. 6-19 and 6-20: SEQ ID NO:269), LMO2 (FIG. 6-21: SEQ ID NO:270), YIPF3 (FIG. 6-22: SEQ ID NO:271), SMN1 (FIG. 6-23: SEQ ID NO:272), CD79B (FIG. 6-24: SEQ ID NO:273), CD44 (FIGS. 6-25 and 6-26: SEQ ID NO:274), CTSC (FIG. 6-27: SEQ ID NO:275), UAP1 (FIG. 6-28: SEQ ID NO:276), PUS7 (FIGS. 6-29 and 6-30: SEQ ID NO:277), RGS13 (FIG. 6-31: SEQ ID NO:278), CD22 (FIGS. 6-32 and 6-33: SEQ ID NO:279), SMN1 (FIG. 6-34: SEQ ID NO:280), and YIPF3 (FIG. 6-35: SEQ ID NO:281).
FIG. 7. Association of multivariate sensitivity index and percent change in tumor sum of the product of diameters (SPD) measurements for 21 patients in Clinical Trial 001. SPD percent change is determined by comparing the smallest post-baseline SPD to baseline SPD. Positive change indicates tumor volume increases, and negative change indicates tumor volume decreases. Weights (coefficients) used for the sensitivity index calculation are shown in Table 14. Larger multivariate sensitivity index values are associated with SPD decreases post-baseline (Sperman's Rho=−0.58; P=0.006).
FIG. 8. Association of BCL6 expression and percent change in SPD measurements for 26 patients with DLBCL. SPD percent change is determined by comparing the smallest post-baseline SPD to baseline SPD. Positive change indicates tumor volume increases, and negative change indicates tumor volume decreases.
DETAILED DESCRIPTION
The present invention is based on the discovery that certain genes (e.g., genes shown in Tables 2-4, 6, 7 and 13) are differentially expressed between B lymphoma cells that are sensitive to anti-CD40 antibody induced cell death and B lymphoma cells that are resistant to anti-CD40 induced cell death. Data from clinical trials described in Example 2 indicate that the expression level of the fourteen genes shown in Table 13 is highly associated with responsiveness to anti-CD40 Ab.1 treatment. Some of the differentially expressed genes between sensitive B lymphoma cells and resistant B lymphoma cells are the CD40 ligand downregulated pathway genes; and some are in the B-cell receptor signaling pathway. Accordingly, expression levels of one or more of these differentially expressed genes can be used for assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to treatment with anti-CD40 antibodies, predicting responsiveness of the subject to treatment with anti-CD40 antibodies, and monitoring treatment/responsiveness in the subject.
A. General Techniques
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, “Molecular Cloning: A Laboratory Manual”, second edition (Sambrook et al., 1989); “Oligonucleotide Synthesis” (M. J. Gait, ed., 1984); “Animal Cell Culture” (R. I. Freshney, ed., 1987); “Methods in Enzymology” (Academic Press, Inc.); “Current Protocols in Molecular Biology” (F. M. Ausubel et al., eds., 1987, and periodic updates); “PCR: The Polymerase Chain Reaction”, (Mullis et al., eds., 1994).
Primers, oligonucleotides and polynucleotides employed in the present invention can be generated using standard techniques known in the art.
Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, N.Y. 1994), and March, Advanced Organic Chemistry Reactions, Mechanisms and Structure 4th ed., John Wiley & Sons (New York, N.Y. 1992), provide one skilled in the art with a general guide to many of the terms used in the present application.
B. Definitions
As used herein, the terms “a subject having a B-cell lymphoma” and “B-cell lymphoma patient” refer to a subject who has been diagnosed with a type of B-cell lymphoma or has been given a probable diagnosis of a type of B-cell lymphoma.
The term “biomarker” or “marker” as used herein refers generally to a molecule, including a gene, protein, carbohydrate structure, or glycolipid, the expression of which in or on a mammalian tissue or cell or secreted can be detected by known methods (or methods disclosed herein) and is predictive or can be used to predict (or aid prediction) for a mammalian cell's or tissue's sensitivity to, and in some embodiments, to predict (or aid prediction) an individual's responsiveness to treatment regimes based on anti-CD40 antibodies.
The term “sample”, as used herein, refers to a composition that is obtained or derived from a subject of interest that contains a cellular and/or other molecular entity that is to be characterized and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics. For example, the phrase “disease sample” and variations thereof refers to any sample obtained from a subject of interest that would be expected or is known to contain the cellular and/or molecular entity that is to be characterized.
By “tissue or cell sample” is meant a collection of similar cells obtained from a tissue of a subject or patient. The source of the tissue or cell sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, or interstitial fluid; cells from any time in gestation or development of the subject. The tissue sample may also be primary or cultured cells or cell lines. Optionally, the tissue or cell sample is obtained from a disease tissue/organ. The tissue sample may contain compounds which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
For the purposes herein a “section” of a tissue sample is meant a single part or piece of a tissue sample, e.g. a thin slice of tissue or cells cut from a tissue sample. It is understood that multiple sections of tissue samples may be taken and subjected to analysis according to the present invention, provided that it is understood that the present invention comprises a method whereby the same section of tissue sample is analyzed at both morphological and molecular levels, or is analyzed with respect to both protein and nucleic acid.
As used herein, a “B-cell lymphoma sample” or a “sample comprising B lymphoma cells” is a tissue or cell sample containing B lymphoma cells from a subject or a patient that have been diagnosed with a type of B-cell lymphoma.
As used herein, method for “aiding assessment” refers to methods that assist in making a clinical determination (e.g., responsiveness of a B-cell lymphoma to treatment with anti-CD40 antibodies), and may or may not be conclusive with respect to the definitive assessment.
A “subject” or an “individual” is a mammal, more preferably a human. Mammals include, but are not limited to, humans, primates, farm animal, sport animals, rodents, and pets (e.g., dogs and cats).
As used herein, a “reference value” can be an absolute value; a relative value; a value that has an upper and/or lower limit; a range of values; an average value; a median value; a mean value; or a value as compared to a particular control or baseline value.
The term “array” or “microarray”, as used herein refers to an ordered arrangement of hybridizable array elements, such as polynucleotide probes (e.g., oligonucleotides) and antibodies, on a substrate. The substrate can be a solid substrate, such as a glass slide, or a semi-solid substrate, such as nitrocellulose membrane. The nucleotide sequences can be DNA, RNA, or any permutations thereof.
“Amplification,” as used herein, generally refers to the process of producing multiple copies of a desired sequence. “Multiple copies” means at least 2 copies. A “copy” does not necessarily mean perfect sequence complementarity or identity to the template sequence. For example, copies can include nucleotide analogs such as deoxyinosine, intentional sequence alterations (such as sequence alterations introduced through a primer comprising a sequence that is hybridizable, but not complementary, to the template), and/or sequence errors that occur during amplification.
Expression/amount of a gene or biomarker in a first sample is at a level “greater than” the level in a second sample if the expression level/amount of the gene or biomarker in the first sample is at least about 1.5×, 1.75×, 2×, 3×, 4×, 5×, 6×, 7×, 8×, 9× or 10× the expression level/amount of the gene or biomarker in the second sample. Expression levels/amounts can be determined based on any suitable criterion known in the art, including but not limited to mRNA, cDNA, proteins, protein fragments and/or gene copy. Expression levels/amounts can be determined qualitatively and/or quantitatively.
“Polynucleotide,” or “nucleic acid,” as used interchangeably herein, refer to polymers of nucleotides of any length, and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the polymer. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. Other types of modifications include, for example, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, cabamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide(s). Further, any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports. The 5′ and 3′ terminal OH can be phosphorylated or substituted with amines or organic capping groups moieties of from 1 to 20 carbon atoms. Other hydroxyls may also be derivatized to standard protecting groups. Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2′-O-methyl-2′-O-allyl, 2′-fluoro- or 2′-azido-ribose, carbocyclic sugar analogs, α-anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and a basic nucleoside analogs such as methyl riboside. One or more phosphodiester linkages may be replaced by alternative linking groups. These alternative linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S(“thioate”), P(S)S (“dithioate”), “(O)NR 2 (“amidate”), P(O)R, P(O)OR′, CO or CH 2 (“formacetal”), in which each R or R′ is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (—O—) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
“Oligonucleotide,” as used herein, generally refers to short, generally single stranded, generally synthetic polynucleotides that are generally, but not necessarily, less than about 200 nucleotides in length. The terms “oligonucleotide” and “polynucleotide” are not mutually exclusive. The description above for polynucleotides is equally and fully applicable to oligonucleotides.
A “primer” is generally a short single stranded polynucleotide, generally with a free 3′-OH group, that binds to a target potentially present in a sample of interest by hybridizing with a target sequence, and thereafter promotes polymerization of a polynucleotide complementary to the target. A “pair of primers” refer to a 5′ primer and a 3′ primer that can be used to amplify a portion of a specific target gene.
The term “3′” generally refers to a region or position in a polynucleotide or oligonucleotide 3′ (downstream) from another region or position in the same polynucleotide or oligonucleotide. The term “5′” generally refers to a region or position in a polynucleotide or oligonucleotide 5′ (upstream) from another region or position in the same polynucleotide or oligonucleotide.
The phrase “gene amplification” refers to a process by which multiple copies of a gene or gene fragment are formed in a particular cell or cell line. The duplicated region (a stretch of amplified DNA) is often referred to as “amplicon.” Usually, the amount of the messenger RNA (mRNA) produced, i.e., the level of gene expression, also increases in the proportion of the number of copies made of the particular gene expressed.
“Detection” includes any means of detecting, including direct and indirect detection.
The term “prediction” is used herein to refer to the likelihood that a patient will respond either favorably or unfavorably to a drug or set of drugs. In one embodiment, the prediction relates to the extent of those responses. In one embodiment, the prediction relates to whether and/or the probability that a patient will survive or improve following treatment, for example treatment with a particular therapeutic agent, and for a certain period of time without disease recurrence. The predictive methods of the invention can be used clinically to make treatment decisions by choosing the most appropriate treatment modalities for any particular patient. The predictive methods of the present invention are valuable tools in predicting if a patient is likely to respond favorably to a treatment regimen, such as a given therapeutic regimen, including for example, administration of a given therapeutic agent or combination, surgical intervention, steroid treatment, etc., or whether long-term survival of the patient, following a therapeutic regimen is likely.
The term “long-term” survival is used herein to refer to survival for at least 1 year, 5 years, 8 years, or 10 years following therapeutic treatment.
“Patient response” can be assessed using any endpoint indicating a benefit to the patient, including, without limitation, (1) inhibition, to some extent, of disease progression, including slowing down and complete arrest; (2) reduction in the number of disease episodes and/or symptoms; (3) reduction in lesional size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of disease cell infiltration into adjacent peripheral organs and/or tissues; (5) inhibition (i.e. reduction, slowing down or complete stopping) of disease spread; (6) relief, to some extent, of one or more symptoms associated with the disorder; (7) increase in the length of disease-free presentation following treatment; and/or (8) decreased mortality at a given point of time following treatment.
The term “antibody” is used in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonal antibodies), multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity or function.
“Antibody fragments” comprise a portion of a full length antibody, generally the antigen binding or variable region thereof. Examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
“Fv” is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. From the folding of these two domains emanate six hypervariable loops (3 loops each from the H and L chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
The term “monoclonal antibody” as used herein refers to an antibody from a population of substantially homogeneous antibodies, i e., the individual antibodies comprising the population are identical and/or bind the same epitope(s), except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts. Such monoclonal antibody typically includes an antibody comprising a polypeptide sequence that binds a target, wherein the target-binding polypeptide sequence was obtained by a process that includes the selection of a single target binding polypeptide sequence from a plurality of polypeptide sequences. For example, the selection process can be the selection of a unique clone from a plurality of clones, such as a pool of hybridoma clones, phage clones or recombinant DNA clones. It should be understood that the selected target binding sequence can be further altered, for example, to improve affinity for the target, to humanize the target binding sequence, to improve its production in cell culture, to reduce its immunogenicity in vivo, to create a multispecific antibody, etc., and that an antibody comprising the altered target binding sequence is also a monoclonal antibody of this invention. In contrast to polyclonal antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. In addition to their specificity, the monoclonal antibody preparations are advantageous in that they are typically uncontaminated by other immunoglobulins. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including, for example, the hybridoma method (e.g., Kohler et al., Nature, 256:495 (1975); Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681, (Elsevier, N.Y., 1981)), recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567), phage display technologies (see, e.g., Clackson et al., Nature, 352:624-628 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991); Sidhu et al., J. Mol. Biol. 338(2):299-310 (2004); Lee et al., J. Mol. Biol. 340(5):1073-1093 (2004); Fellouse, Proc. Nat. Acad. Sci. USA 101(34):12467-12472 (2004); and Lee et al. J. Immunol. Methods 284(1-2):119-132 (2004), and technologies for producing human or human-like antibodies in animals that have parts or all of the human immunoglobulin loci or genes encoding human immunoglobulin sequences (see, e.g., WO 1998/24893; WO 1996/34096; WO 1996/33735; WO 1991/10741; Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90:2551 (1993); Jakobovits et al., Nature, 362:255-258 (1993); Bruggemann et al., Year in Immuno., 7:33 (1993); U.S. Pat. Nos. 5,545,806; 5,569,825; 5,591,669 (all of GenPharm); 5,545,807; WO 1997/17852; U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016; Marks et al., Bio/Technology, 10: 779-783 (1992); Lonberg et al., Nature, 368: 856-859 (1994); Morrison, Nature, 368: 812-813 (1994); Fishwild et al., Nature Biotechnology, 14: 845-851 (1996); Neuberger, Nature Biotechnology, 14: 826 (1996); and Lonberg and Huszar, Intern. Rev. Immunol., 13: 65-93 (1995).
The monoclonal antibodies herein specifically include “chimeric” antibodies. “Chimeric” antibodies (immunoglobulins) have a portion of the heavy and/or light chain identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984)). Humanized antibody as used herein is a subset of chimeric antibodies.
“Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies which contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient or acceptor antibody) in which hypervariable region residues of the recipient are replaced by hypervariable region residues from a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues which are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance such as binding affinity. Generally, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence although the FR regions may include one or more amino acid substitutions that improve binding affinity. The number of these amino acid substitutions in the FR are typically no more than 6 in the H chain, and in the L chain, no more than 3. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al., Nature 321:522-525 (1986); Reichmann et al., Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol. 2:593-596 (1992).
A “human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human and/or has been made using any of the known techniques for making human antibodies. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
An “affinity matured” antibody is one with one or more alterations in one or more CDRs/HVRs thereof which result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s). Preferred affinity matured antibodies will have nanomolar or even picomolar affinities for the target antigen. Affinity matured antibodies are produced by procedures known in the art. Marks et al. Bio/Technology 10:779-783 (1992) describes affinity maturation by VH and VL domain shuffling. Random mutagenesis of CDR/HVR and/or framework residues is described by: Barbas et al. Proc Nat. Acad. Sci, USA 91:3809-3813 (1994); Schier et al. Gene 169:147-155 (1995); Yelton et al. J. Immunol. 155:1994-2004 (1995); Jackson et al., J. Immunol. 154(7):3310-9 (1995); and Hawkins et al, J. Mol. Biol. 226:889-896 (1992).
The term “Fc region” is used to define the C-terminal region of an immunoglobulin heavy chain which may be generated by papain digestion of an intact antibody. The Fc region may be a native sequence Fc region or a variant Fc region. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at about position Cys226, or from about position Pro230, to the carboxyl-terminus of the Fc region. The Fc region of an immunoglobulin generally comprises two constant domains, a CH2 domain and a CH3 domain, and optionally comprises a CH4 domain. By “Fc region chain” herein is meant one of the two polypeptide chains of an Fc region.
Antibody “effector functions” refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody, and vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
“Antibody-dependent cell-mediated cytotoxicity” or “ADCC” refers to a form of cytotoxicity in which secreted Ig bound onto Fc receptors (FcRs) present on certain cytotoxic cells (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) enable these cytotoxic effector cells to bind specifically to an antigen-bearing target cell and subsequently kill the target cell with cytotoxins. The antibodies “arm” the cytotoxic cells and are absolutely required for such killing. The primary cells for mediating ADCC, NK cells, express FcγRIII only, whereas monocytes express FcγRI, FcγRII and FcγRIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991). To assess ADCC activity of a molecule of interest, an in vitro ADCC assay, such as that described in U.S. Pat. No. 5,500,362 or 5,821,337 or Presta U.S. Pat. No. 6,737,056 may be performed. Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. PNAS (USA) 95:652-656 (1998).
“Treating” or “treatment” or “alleviation” refers to therapeutic treatment wherein the object is to slow down (lessen) if not cure the targeted pathologic condition or disorder or prevent recurrence of the condition. A subject is successfully “treated” for the B cell malignancy if, after receiving a therapeutic amount of a CD40 binding antibody, the subject shows observable and/or measurable reduction in or absence of one or more signs and symptoms of the particular disease. For example, significant reduction in the number of cancer cells or absence of the cancer cells; reduction in the tumor size; inhibition (i.e., slow to some extent and preferably stop) of tumor metastasis; inhibition, to some extent, of tumor growth; increase in length of remission, and/or relief to some extent, one or more of the symptoms associated with the specific cancer; reduced morbidity and mortality, and improvement in quality of life issues. Reduction of the signs or symptoms of a disease may also be felt by the patient. Treatment can achieve a complete response, defined as disappearance of all signs of cancer, or a partial response, wherein the size of the tumor is decreased, preferably by more than 50 percent, more preferably by 75%. A patient is also considered treated if the patient experiences stable disease. In one criterion, the antibodies of the invention achieve >95% peripheral blood B cell depletion and the B cells return to 25% of baseline. In some embodiments, treatment with the anti-CD40 antibodies is effective to result in the cancer patients being progression-free in the cancer 3 months after treatment, preferably 6 months, more preferably one year, even more preferably 2 or more years post treatment. These parameters for assessing successful treatment and improvement in the disease are readily measurable by routine procedures familiar to a physician of appropriate skill in the art.
The term “non-Hodgkin's lymphoma” or “NHL”, as used herein, refers to a cancer of the lymphatic system other than Hodgkin's lymphomas. Hodgkin's lymphomas can generally be distinguished from non-Hodgkin's lymphomas by the presence of Reed-Sternberg cells in Hodgkin's lymphomas and the absence of said cells in non-Hodgkin's lymphomas.
An “effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. A “therapeutically effective amount” of a therapeutic agent may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the therapeutic agent are outweighed by the therapeutically beneficial effects. A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
The term “housekeeping gene” refers to a group of genes that codes for proteins whose activities are essential for the maintenance of cell function. These genes are typically similarly expressed in all cell types.
By “correlate” or “correlating” is meant comparing, in any way, the performance and/or results of a first analysis or protocol with the performance and/or results of a second analysis or protocol. For example, one may use the results of a first analysis or protocol in carrying out a second protocols and/or one may use the results of a first analysis or protocol to determine whether a second analysis or protocol should be performed. With respect to the embodiment of gene expression analysis or protocol, one may use the results of the gene expression analysis or protocol to determine whether a specific therapeutic regimen should be performed.
The word “label” when used herein refers to a compound or composition which is conjugated or fused directly or indirectly to a reagent such as a nucleic acid probe or an antibody and facilitates detection of the reagent to which it is conjugated or fused. The label may itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
As used herein, “a”, “an”, and “the” can mean singular or plural (i.e., can mean one or more) unless indicated otherwise.
C. Methods of the Invention
The invention provides methods for assessing or aiding assessment of responsiveness of a subject having a B-cell lymphoma to treatment with an anti-CD40 antibody. The invention also provides methods for predicting responsiveness or monitoring treatment/responsiveness to an anti-CD40 antibody treatment in a subject having a B-cell lymphoma. The invention provides methods for selecting a subject having a B-cell lymphoma suitable for treatment with an anti-CD40 antibody and following up with an anti-CD40 antibody treatment. In some embodiments, the methods comprise measuring expression level of one or more marker genes in any of Tables 2-4, 6, 7, and 13 in a sample comprising B lymphoma cells obtained from the subject; and predicting, assessing, or aiding assessment of responsiveness of the subject to an anti-CD40 antibody treatment based on the measure expression level of said one or more marker genes. In some embodiments, the methods comprise comparing a measured expression level of at least one marker gene in any of Tables 2-4, 6, 7, and 13 in a B-cell lymphoma sample from the subject to a reference level for the respective marker gene.
The methods of the present invention are useful for clinicians to identify patients with B-cell lymphoma for treatment with an anti-CD40 antibody, aiding in patient selection during the course of development of anti-CD40 antibody therapy, prediction of likelihood of success when treating an individual patient with a particular treatment regimen, in assessing and monitoring disease progression, in monitoring treatment efficacy, and in determining prognosis for individual patients. Any of these embodiments are included in this invention.
In some embodiments, the B-cell lymphoma is non-Hodgkin's lymphoma (NHL), including, but is not limited to, follicular lymphoma, relapsed follicular lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, lymphoplasmacytic lymphoma, mycosis fungoides/Sezary syndrome, splenic marginal zone lymphoma, and diffuse large B-cell lymphoma.
In some embodiments, the B-cell lymphoma is indolent. In some embodiments, the B-cell lymphoma is aggressive. In some embodiments, the B-cell lymphoma is highly aggressive. In some embodiments, the indolent B-cell lymphoma is follicular lymphoma, marginal zone lymphoma, or small lymphocytic lymphoma. In some embodiments, the indolent B-cell lymphoma is follicular lymphoma.
Marker Genes
The expression level of one or more of the marker genes in a B-cell lymphoma sample relative a reference level may be used in the methods of the invention, such as to predict, assess or aid assessment of responsiveness of the B-cell lymphoma to treatment with an anti-CD40 antibody.
Genes that are differentially expressed (statistically significantly increased or decreased) in anti-CD40 antibody sensitive NHL cell lines as compared to resistant NHL cell lines are shown in Tables 2-4, 6 and 7. “Anti-CD40 antibody sensitive cells” are cells having an IC25 value less than 0.4 μg/ml in reduction of cell viability by an anti-CD40 antibody tested as described in Example 1. “Anti-CD40 resistant cells” are cells having an IC25 value greater than 1 μg/ml in reduction in cell viability as tested in Example 1. Some of the genes in Tables 2-4, 6 and 7 are in the CD40 ligand downregulated pathway (for example, VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, and POU2AF1); and some of the genes in the tables are in the B-cell receptor signaling pathway (for example, CD22, RGS13, and MEF2B). Further, association of the expression level of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 (Table 13) has been confirmed by clinical trials described in Example 2. Expression levels of one or more of these genes are used in the methods of the invention. In some embodiments, expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least twenty, at least twenty five, or at least thirty genes are used in the methods of the invention.
In some embodiments, expression levels of one or more of genes selected from the group consisting of VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, and MEF2B are measured and/or used. In some embodiments, expression levels of one or more of genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are measured and/or used. In some embodiments, expression levels of at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, or fourteen of these genes are measured and/or used. In some embodiments, expression levels of CD22, CD40, and BCL6 are measured and/or used. In some embodiments, expression levels of CD40, RGS13, CD22, BTG2, IGF1R, and CD44 are measured and/or used. In some embodiments, expression levels of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are measured and/or used. In some embodiments, expression levels of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or fifteen of genes in Table 7 or Table 13 are measured and/or used.
Genes (including sequences) identified in Tables 2-4, 6, 7 and 13 are known in the art. For example, the examples of Gene Bank accession numbers for human genes are VNN2 (NM004665; NM078488; AJ132100; D89974; BC064641; CR609799; BC126145; BC126147; and AB026705); RGS13 (NM002927; NM144766; BT006929; BC056866; AY562947; CR536532; CR610389; CR599001; BC016667; AF493935; BC036950; and AF030107); CD22 (NM001771; AK026467; BC109306; BC109307; AK225694; AK225625; X52785; and X59350); LRRC8A (AY143166; BC051322; AK123611; AY358286; NM019594; XM026998; AK001199; AB037858; CR619692; CR619448; AK024649; BC000775; AK027495; and AK074723); CD40 (NM001250; NM152854; BC064518; AY225405; CR619622; CR608994; CR605787; AB209660; AK222896; AJ300189; BT019901; and BC012419); IFITM1 (NM003641; BC000897; BT007173; BT009859; CR456894; CR541874; CR604902; X57351; X84958; NM006435; BC009696; X02490; and J04164); SMN1 (NM000344; BC062723; CR611445; CR593735; BC000908; NM022874; BC015308; and U18423); PRKCA (NM002737; AB209475; BC109274; BC109273; AF035594; BC053321; BX648954; AK125425; BC062759; BC071767; BC103691; BC101403; BC107592; AY633609; BC122530; BC015855; AF086287; AF035595; M22199; and X52479); EPDR1 (DQ914439; AY027862; NM017549; AJ250475; AF202051; CR624676; CR596656; NM016616; BC000686; BC018299; AF305596; and BC036816); PRPSAP2 (NM002767; AB007851; BX648850; AK126398; CR457082; BC101672; BC101670; and BC106050); IGF1R(NM000875; NM015883; AY429545; CR624013; BC078157; BC088377; BC107089; BC111046; BC113610; BC113612; BC010607; X04434 M24599; and U09023); BTG2 (NM006763; CR606002; CR604962; CR595352; CR591042; BC105948; BC105949; U72649; and Y09943); LMO2 (BC042426; NM005574; BC073973; AK127915; CR625714; CR614368; CR604507; AF257211; BC034041; BC035607; and X61118); YIPF3 (AL050274; AK000946; CR533541; CR623137; CR622890; CR622532; CR621993; CR619816; CR619437; CR619054; CR618212; CR616987; CR616384; CR615623; CR615153; CR615118; CR612415; CR611748; CR611260; CR610983; CR610470; CR607768; CR606024; CR603408; CR603202; CR602267; CR601987; CR599615; CR598162; CR597677; CR596581; CR596249; CR595236; CR592266; CR590752; CR590349; NM015388; AK021433; AK021655; AK022757; BC019297; and AF162672); and BCL6 (NM001706; NM138931; BX649185; U00115; BC142705; BC146796; BC150184; AL713713; AK090890; AL832990; and Z21943).
The nucleic acid sequence of some of the genes referenced in Tables 2-4, 6, 7 and 13 are shown in FIG. 6 (6-1 to 6-35).
Reference Levels
The measured expression level of one or more marker genes in a B-cell lymphoma sample is compared to a reference level. In some embodiments, the reference level is the expression level of a gene the expression level of which does not change (does not change significantly) among different type of B-cell lymphomas, for example, between B-cell lymphoma sensitive to anti-CD40 antibody and B-cell lymphoma resistant to anti-CD40 antibody. In some embodiments, expression levels of one or more housekeeping genes shown in Table 8 are used as reference levels. In some embodiments, expression levels of one or more housekeeping genes shown in Table 9 are used as reference levels.
In some embodiments, the measured expression level of the marker gene is normalized using the reference level. In some embodiments, the normalized expression level of the marker gene is calculated as a ratio of or difference between the marker gene and reference expression levels, on the original or on a log scale, respectively.
The reference genes in Table 8 and Table 9 were selected as specific normalizing counterparts to the marker genes in Table 4. Reference genes were selected for high mean expression and low variance in B cell lymphoma samples. In addition, reference genes were selected to have similar variance between replicated expression measurements of individual cell lines relative to variance between expression measurements of biologically distinct cell lines. In addition, reference genes were selected to have low statistical association with one or more markers in Table 4.
In some embodiments, the reference level is a measured expression level of the marker gene in a different B-cell lymphoma sample. In some embodiments, the different B cell lymphoma sample comprises B lymphoma cells that are resistant to an anti-CD40 antibody induced cell death.
In some embodiments, the reference level is determined based on the expression level of the corresponding marker gene in samples comprising B lymphoma cells from subjects having tumor volume increased after the anti-CD40 antibody treatment and/or having tumor volume decreased after the anti-CD40 antibody treatment. In some embodiments, the samples from subjects for reference level determination comprise the same type of B lymphoma cells as the sample from the subject whose responsiveness to the anti-CD40 antibody treatment is predicted or assessed. In some embodiments, the same method (e.g., qRT-PCR) and/or reagents (e.g., primers and probes) are used for measuring expression level of the marker genes in the sample and measuring expression level of the corresponding marker genes in the reference samples.
TABLE 8
SCR. IC25. GCB. SCR EXT.
VarW. vscr. anti-CD40. anti-CD40. anti-CD40. anti-CD40.
Probe symb VarB mean var vscr rank P. Min Ab.1 Ab.1 Ab.1 Ab.1
202521_at CTCF 0.02 10.61 0.19 −3.81 5079 0.020543 0.896744 0.758931 0.927787 0.285815
201949_x_at CAPZB 0.04 11.78 0.33 7.92 300 0.363476 0.5627 0.9554 0.3785 0.3635
201588_at TXNL1 0.01 13.00 0.29 −2.39 3182 2.49E−09 0.2422 0.5231 0.2540 0.1104
201070_x_at SF3B1 0.20 9.46 0.23 −3.78 5023 0.089689 0.1715 0.1517 0.2230 0.5294
209180_at RABGGTB 0.23 10.80 0.40 −2.89 3693 0.001233 0.9074 0.9214 0.7339 0.1495
AFFX- ACTB 0.03 14.02 0.53 0.48 2039 0.144577 0.6074 0.9584 0.2415 0.4461
HSAC07/
X00351_5_at
201891_s_at B2M 0.13 14.67 0.22 1.98 1919 0.010118 0.2646 0.1011 0.4501 0.0392
FFX- GAPDH 0.59 14.78 0.04 2.95 1850 0.000944 0.7089 0.7244 0.9014 0.3096
HUMGAPDH/
M33197_5_at
202605_at GUSB 0.05 10.52 0.65 −3.44 4415 6.73E−05 0.0096 0.0104 0.0053 0.0885
202854_at HPRT1 0.03 12.92 0.30 −1.90 2773 2.64E−05 0.1297 0.2069 0.0532 0.5541
200737_at PGK1 0.02 12.20 0.46 −2.75 3533 0.000307 0.0777 0.3535 0.0719 0.6473
201293_x_at PPIA 0.60 14.99 0.02 3.98 1731 0.065694 0.1406 0.3579 0.1735 0.6190
201033_x_at RPLP0 0.62 15.20 0.01 4.16 1709 0.066741 0.0667 0.1150 0.1081 0.7451
203135_at TBP 0.06 8.29 0.19 −16.66 21417 0.001289 0.6978 0.7904 0.8630 0.2849
207332_s_at TFRC 0.06 12.50 1.16 −0.82 2311 5.66E−06 0.1391 0.0963 0.1051 0.1710
226131_s_at RPS16 0.68 15.60 0.01 15.24 1 0.4182 0.6946 0.6783 0.9425 0.4182
1553567_s_at ATP13A5 0.53 15.77 0.04 15.10 2 0.2744 0.3205 0.5881 0.2744 0.8039
213477_x_at EEF1A1 0.80 15.71 0.02 14.94 3 0.2716 0.3490 0.5611 0.2716 0.9425
229563_s_at RPL10A 0.65 15.08 0.02 14.64 4 0.2266 0.3258 0.2266 0.6668 0.7720
203107_x_at RPS2 0.75 15.37 0.01 14.55 5 0.2635 0.4033 0.5834 0.2635 0.6664
213614_x_at EEF1A1 0.51 16.11 0.02 14.47 6 0.2273 0.4168 0.5721 0.2273 0.6765
204892_x_at EEF1A1 0.55 15.29 0.02 14.46 7 0.3353 0.7883 0.7755 0.5296 0.7598
212391_x_at RPS3A 0.78 15.00 0.01 14.34 8 0.2519 0.3159 0.6319 0.2519 0.3350
211542_x_at RPS10 0.59 15.11 0.02 14.31 9 0.2000 0.8313 0.9604 0.7117 0.7029
213583_x_at EEF1A1 0.66 15.26 0.04 14.29 10 0.2172 0.4132 0.7604 0.2172 0.8064
200819_s_at RPS15 0.54 15.00 0.05 13.99 11 0.3700 0.6401 0.7339 0.8220 0.7939
200095_x_at FLJ20294 0.60 15.29 0.02 13.98 12 0.3400 0.7334 0.5003 0.4045 0.4757
224585_x_at ACTG1 0.49 14.73 0.06 13.96 13 0.4788 0.9612 0.7590 0.4788 0.5097
213414_s_at RPS19 0.49 15.19 0.02 13.95 14 0.3134 0.6110 0.5909 0.3134 0.9180
1553538_s_at NA 0.33 15.24 0.24 13.94 15 0.5473 0.6181 0.5473 0.9966 0.9360
200032_s_at RPL9 0.61 15.30 0.01 13.80 16 0.2652 0.7969 0.6658 0.8910 0.9033
200063_s_at NPM1 0.68 15.34 0.02 13.78 17 0.2634 0.6557 0.7122 0.2634 0.9201
213890_x_at RPS16 0.42 15.02 0.01 13.68 18 0.2333 0.2936 0.2333 0.3297 0.2718
212734_x_at RPL13 0.46 14.92 0.03 13.66 19 0.2300 0.8232 0.6720 0.4503 0.7004
211983_x_at ACTG1 0.40 14.83 0.06 13.54 20 0.4100 0.9680 0.7211 0.4205 0.7919
213801_x_at RPSA 0.61 15.01 0.05 13.53 21 0.2661 0.4603 0.7140 0.2661 0.4003
202649_x_at RPS19 0.33 15.03 0.03 13.44 22 0.3172 0.5861 0.5086 0.3172 0.9400
221607_x_at ACTG1 0.41 14.73 0.06 13.38 23 0.2715 0.9680 0.6637 0.3927 0.6126
212988_x_at ACTG1 0.45 14.53 0.06 13.31 24 0.3553 0.9075 0.6394 0.3553 0.7217
208929_x_at RPL13 0.40 14.75 0.02 13.25 25 0.3500 0.3583 0.7912 0.9760 0.6997
200689_x_at EEF1G 0.64 14.25 0.03 13.21 26 0.2100 0.9324 0.8163 0.8508 0.3667
211345_x_at EEF1G 0.54 14.23 0.03 13.21 27 0.2200 0.9444 0.8022 0.7118 0.3901
211970_x_at ACTG1 0.46 14.51 0.09 13.18 28 0.3072 0.7347 0.8427 0.7238 0.5534
211995_x_at ACTG1 0.35 14.61 0.10 13.14 29 0.3981 0.5436 0.9959 0.9161 0.3981
200089_s_at RPL4 0.29 15.28 0.04 13.09 30 0.2068 0.4500 0.6581 0.5132 0.5295
200024_at RPS5 0.57 14.61 0.03 13.09 31 0.2000 0.7753 0.8060 0.5846 0.9469
201550_x_at ACTG1 0.33 14.50 0.10 13.04 32 0.3356 0.5966 0.9624 0.8531 0.4378
AFFX-r2-P1- NA 0.28 15.23 0.12 13.00 33 0.4500 0.9518 0.5889 0.7244 0.8836
cre-3_at
200003_s_at RPL28 0.14 15.12 0.04 12.93 34 0.5687 0.9905 0.6582 0.9539 0.5687
212363_x_at ACTG1 0.33 14.18 0.12 12.81 35 0.4219 0.6539 0.8152 0.8079 0.4254
221775_x_at EVI1 0.28 14.55 0.05 12.78 36 0.2391 0.9589 0.8109 0.4979 0.8711
208768_x_at RPL22 0.30 14.56 0.05 12.78 37 0.2858 0.9577 0.8867 0.4568 0.9964
212191_x_at LOC388344 0.18 14.84 0.05 12.77 38 0.2500 0.9777 0.8542 0.3553 0.9844
200021_at CFL1 0.50 13.77 0.02 12.77 39 0.2775 0.8529 0.8339 0.5283 0.2775
208517_x_at BTF3 0.33 14.54 0.02 12.56 40 0.2513 0.7046 0.7417 0.9434 0.2954
211956_s_at EIF1 0.16 15.12 0.08 12.50 41 0.2756 0.2756 0.3283 0.6567 0.4596
214351_x_at RPL13 0.44 14.01 0.03 12.36 42 0.2703 0.4829 0.9230 0.9173 0.4119
224731_at HMGB1 0.11 14.37 0.17 12.35 43 0.3496 0.4679 0.9363 0.4219 0.3496
234512_x_at LOC388474 0.25 13.55 0.04 12.35 44 0.5910 0.9435 0.5910 0.9578 0.6021
220960_x_at RPL22 0.28 14.20 0.02 12.28 45 0.5585 0.7556 0.8571 0.7995 0.9640
221791_s_at CCDC72 0.45 14.33 0.03 12.22 46 0.2692 0.5460 0.8746 0.4059 0.2692
216438_s_at TMSB4X 0.04 15.34 1.15 12.02 47 0.2086 0.3130 0.2155 0.2086 0.8821
201030_x_at LDHB 0.22 14.68 0.05 11.91 48 0.3032 0.4740 0.8098 0.5684 0.3032
AFFX-CreX- NA 0.27 14.50 0.19 11.83 49 0.4700 0.9276 0.5873 0.7234 0.9267
3_at
200715_x_at RPL13A 0.26 13.87 0.12 11.70 50 0.3000 0.8556 0.3818 0.6143 0.4458
AFFX-CreX- NA 0.15 14.64 0.27 11.59 51 0.3900 0.9872 0.6546 0.5814 0.7754
5_at
222976_s_at TPM3 0.04 14.13 0.09 11.54 52 0.3646 0.3786 0.7883 0.3646 0.5240
210466_s_at SERBP1 0.52 13.90 0.07 11.51 53 0.2326 0.3230 0.2326 0.2545 0.8323
225413_at USMG5 0.07 13.78 0.15 11.49 54 0.3239 0.9696 0.5515 0.8338 0.3239
221691_x_at NPM1 0.10 15.00 0.07 11.44 55 0.5097 0.8965 0.7627 0.5097 0.7686
229353_s_at NUCKS1 0.07 13.62 0.21 11.21 56 0.6703 0.7457 0.6703 0.7602 0.8020
1555730_a_at CFL1 0.04 14.01 0.30 11.17 57 0.4996 0.9337 0.7560 0.4996 0.5768
200966_x_at ALDOA 0.09 14.02 0.11 11.09 58 0.2409 0.2409 0.5526 0.4352 0.8701
224654_at DDX21 0.06 13.50 0.13 11.07 59 0.6759 0.8439 0.8720 0.7694 0.6759
224944_at TMPO 0.05 13.48 0.14 10.98 60 0.2455 0.3257 0.4478 0.3876 0.2455
222985_at YWHAG 0.04 13.53 0.15 10.86 61 0.3506 0.7800 0.3506 0.9581 0.9505
1555837_s_at POLR2B 0.07 13.18 0.16 10.85 62 0.3371 0.8399 0.3612 0.6857 0.3371
209026_x_at TUBB 0.07 13.60 0.24 10.73 63 0.2100 0.6957 0.4642 0.7072 0.3910
238199_x_at LOC440552 0.56 11.52 0.17 10.69 64 0.2720 0.3736 0.9156 0.2720 0.6534
217807_s_at GLTSCR2 0.07 13.62 0.19 10.61 65 0.5757 0.8314 0.7256 0.7767 0.5757
242131_at LOC440552 0.53 11.20 0.10 10.42 66 0.5733 0.7746 0.5733 0.7302 0.9388
222980_at RAB10 0.12 12.27 0.13 10.40 67 0.2461 0.7382 0.9132 0.2877 0.2461
234339_s_at GLTSCR2 0.58 11.32 0.29 10.39 68 0.6277 0.7136 0.9772 0.8445 0.6277
1554678_s_at HNRPDL 0.04 13.21 0.22 10.39 69 0.3095 0.3381 0.3095 0.6767 0.5390
200893_at SFRS10 0.12 13.68 0.06 10.38 70 0.3885 0.5944 0.8186 0.6001 0.3885
223105_s_at TMEM14C 0.02 13.54 0.16 10.35 71 0.6699 0.6699 0.8055 0.8667 0.9663
224579_at SLC38A1 0.02 13.49 0.20 10.21 72 0.2496 0.7799 0.3541 0.3506 0.2496
1558678_s_at MALAT1 0.16 12.46 0.89 10.21 73 0.4393 0.9362 0.8914 0.4393 0.9347
223096_at NOP5/NOP58 0.03 13.04 0.13 10.13 74 0.6162 0.6964 0.8240 0.6162 0.6685
224567_x_at MALAT1 0.11 12.50 0.69 10.10 75 0.4566 0.9218 0.9662 0.4566 0.7071
226385_s_at C7orf30 0.03 12.99 0.26 10.02 76 0.6285 0.6285 0.9109 0.7478 0.8336
213011_s_at TPI1 0.04 13.56 0.18 9.96 77 0.2442 0.3471 0.6334 0.5709 0.4333
225892_at IREB2 0.10 12.08 0.21 9.94 78 0.4034 0.8084 0.9066 0.6860 0.4034
231896_s_at DENR 0.03 12.80 0.14 9.93 79 0.2977 0.6041 0.7701 0.2977 0.4713
201114_x_at PSMA7 0.12 12.78 0.15 9.87 80 0.4093 0.5862 0.5983 0.8588 0.4093
208738_x_at SUMO2 0.17 14.07 0.02 9.87 81 0.2055 0.4579 0.4606 0.3408 0.2055
224592_x_at HP1BP3 0.13 11.74 0.15 9.86 82 0.6319 0.6899 0.6319 0.8361 0.8069
224935_at EIF2S3 0.03 13.01 0.35 9.86 83 0.2694 0.3291 0.6816 0.2694 0.3207
224736_at CCAR1 0.10 11.79 0.09 9.86 84 0.5647 0.8733 0.9743 0.7364 0.5647
224593_at ZNF664 0.20 11.63 0.37 9.85 85 0.4300 0.5453 0.8490 0.4300 0.9881
224714_at MKI67IP 0.07 12.26 0.23 9.83 86 0.3898 0.8170 0.7194 0.3898 0.5026
223705_s_at GPBP1 0.05 12.26 0.12 9.79 87 0.6059 0.9591 0.9834 0.6059 0.9781
1553575_at NA 0.04 12.71 0.40 9.76 88 0.2247 0.3970 0.3953 0.2247 0.4525
224591_at HP1BP3 0.04 12.57 0.24 9.72 89 0.6293 0.6293 0.6998 0.7775 0.9947
202690_s_at SNRPD1 0.07 13.90 0.13 9.70 90 0.5018 0.7715 0.5905 0.5018 0.5865
223034_s_at C1orf43 0.02 13.12 0.16 9.70 91 0.4517 0.6653 0.4517 0.7044 0.9095
224376_s_at C20orf24 0.06 12.21 0.23 9.69 92 0.5630 0.9644 0.9137 0.8592 0.5630
AFFX-r2-Ec- NA 0.14 14.01 0.48 9.67 93 0.3700 0.8588 0.8057 0.4071 0.4774
bioD-3_at
201277_s_at HNRPAB 0.04 13.17 0.18 9.66 94 0.3203 0.8462 0.3900 0.3789 0.3203
228273_at NA 0.03 12.65 0.19 9.66 95 0.5447 0.5447 0.6935 0.9994 0.8749
202077_at NDUFAB1 0.06 13.06 0.08 9.65 96 0.2839 0.9323 0.6388 0.6981 0.2839
224561_s_at MORF4L1 0.04 12.46 0.18 9.64 97 0.6517 0.9637 0.6517 0.8271 0.7722
211623_s_at FBL 0.04 13.89 0.16 9.63 98 0.4800 0.5149 0.9574 0.8996 0.9424
212626_x_at HNRPC 0.08 13.05 0.14 9.62 99 0.2260 0.5906 0.5146 0.3161 0.4689
229128_s_at ANP32E 0.03 12.72 0.39 9.61 100 0.4422 0.6538 0.8542 0.4422 0.9196
TABLE 9
VarW. vscr. SCR. IC25. GCB. SCR EXT.
Probe symb.gse VarB mean var vscr rank P. Min anti-CD40 anti-CD40 anti-CD40 anti-CD40
226131_s_at RPS16 0.68 15.60 0.01 15.24 1 0.4182 0.6946 0.6783 0.9425 0.4182
1553567_s_at ATP13A5 0.53 15.77 0.04 15.10 2 0.2744 0.3205 0.5881 0.2744 0.8039
213477_x_at EEF1A1 0.80 15.71 0.02 14.94 3 0.2716 0.3490 0.5611 0.2716 0.9425
211542_x_at RPS10 0.59 15.11 0.02 14.31 9 0.2000 0.8313 0.9604 0.7117 0.7029
200095_x_at FLJ20294 0.60 15.29 0.02 13.98 12 0.3400 0.7334 0.5003 0.4045 0.4757
224585_x_at ACTG1 0.49 14.73 0.06 13.96 13 0.4788 0.9612 0.7590 0.4788 0.5097
213414_s_at RPS19 0.49 15.19 0.02 13.95 14 0.3134 0.6110 0.5909 0.3134 0.9180
200032_s_at RPL9 0.61 15.30 0.01 13.80 16 0.2652 0.7969 0.6658 0.8910 0.9033
200063_s_at NPM1 0.68 15.34 0.02 13.78 17 0.2634 0.6557 0.7122 0.2634 0.9201
212734_x_at RPL13 0.46 14.92 0.03 13.66 19 0.2300 0.8232 0.6720 0.4503 0.7004
200689_x_at EEF1G 0.64 14.25 0.03 13.21 26 0.2100 0.9324 0.8163 0.8508 0.3667
200024_at RPS5 0.57 14.61 0.03 13.09 31 0.2000 0.7753 0.8060 0.5846 0.9469
200003_s_at RPL28 0.14 15.12 0.04 12.93 34 0.5687 0.9905 0.6582 0.9539 0.5687
221775_x_at EVI1 0.28 14.55 0.05 12.78 36 0.2391 0.9589 0.8109 0.4979 0.8711
208768_x_at RPL22 0.30 14.56 0.05 12.78 37 0.2858 0.9577 0.8867 0.4568 0.9964
212191_x_at LOC388344 0.18 14.84 0.05 12.77 38 0.2500 0.9777 0.8542 0.3553 0.9844
200021_at CFL1 0.50 13.77 0.02 12.77 39 0.2775 0.8529 0.8339 0.5283 0.2775
208517_x_at BTF3 0.33 14.54 0.02 12.56 40 0.2513 0.7046 0.7417 0.9434 0.2954
211956_s_at EIF1 0.16 15.12 0.08 12.50 41 0.2756 0.2756 0.3283 0.6567 0.4596
224731_at HMGB1 0.11 14.37 0.17 12.35 43 0.3496 0.4679 0.9363 0.4219 0.3496
234512_x_at LOC388474 0.25 13.55 0.04 12.35 44 0.5910 0.9435 0.5910 0.9578 0.6021
221791_s_at CCDC72 0.45 14.33 0.03 12.22 46 0.2692 0.5460 0.8746 0.4059 0.2692
216438_s_at TMSB4X 0.04 15.34 1.15 12.02 47 0.2086 0.3130 0.2155 0.2086 0.8821
201030_x_at LDHB 0.22 14.68 0.05 11.91 48 0.3032 0.4740 0.8098 0.5684 0.3032
222976_s_at TPM3 0.04 14.13 0.09 11.54 52 0.3646 0.3786 0.7883 0.3646 0.5240
210466_s_at SERBP1 0.52 13.90 0.07 11.51 53 0.2326 0.3230 0.2326 0.2545 0.8323
225413_at USMG5 0.07 13.78 0.15 11.49 54 0.3239 0.9696 0.5515 0.8338 0.3239
221691_x_at NPM1 0.10 15.00 0.07 11.44 55 0.5097 0.8965 0.7627 0.5097 0.7686
Measuring Expression Level
The methods disclosed herein provide methods to examine expression level of one or more of these marker genes in a lymphoma sample (e.g., B-cell lymphoma sample) relative a reference level. The methods and assays include those which examine expression of marker genes such as one or more of those listed in any of Tables 2-4, 6, 7 and 13. Expression levels may be measured at mRNA level and/or protein level.
The invention provides methods for measuring levels of expression from a mammalian tissue or cells sample (such as cells and/or tissues associated with B-cell lymphoma). For example, for obtaining patient samples, H&E staining is carried out and used as a guide for tissue macrodissection to enrich for tumor content. The sample can be obtained by a variety of procedures known in the art including, but is not limited to surgical excision, aspiration or biopsy. The sample may be fresh or frozen. In some embodiments, the sample is fixed and embedded in paraffin or the like. In the methods, a mammalian tissue or cell sample is obtained and examined for expression of one or more biomarkers. The methods may be conducted in a variety of assay formats, including assays detecting mRNA expression, enzymatic assays detecting presence of enzymatic activity, and immunohistochemistry assays. Determination of expression of such biomarkers in said tissues or cells will be predictive that such tissues or cells will be sensitive/responsive to treatment with an anti-CD40 antibody.
As discussed below, expression of various biomarkers in a sample can be analyzed by a number of methodologies, many of which are known in the art and understood by the skilled artisan, including but not limited to, microarray (gene and/or tissue array analysis), in situ hybridization, Northern analysis, PCR analysis of mRNAs, immunohistochemical and/or Western analysis, quantitative blood based assays (as for example Serum ELISA) (to examine, for example, levels of protein expression), and/or biochemical enzymatic activity assays. Typical protocols for evaluating the status of genes and gene products are found, for example in Ausubel et al. eds., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting) and 18 (PCR Analysis). The protocols below relating to detection of particular biomarkers, such as those listed in Tables 2-4, 6, 7 and 13, in a sample are provided for illustrative purposes.
In some embodiments, the methods of the invention further include protocols which examine the presence and/or expression of mRNAs, such as mRNAs of genes listed in any of Tables 2-4, 6, 7 and 13, in a tissue or cell sample. In some embodiments, expression of various biomarkers in a sample may be analyzed by microarray technologies, which examine or detect mRNAs, such as mRNAs in any of Tables 2-4, 6, 7 and 13, in a tissue or cell sample. Using nucleic acid microarrays, test and control mRNA samples from test and control tissue samples are reverse transcribed and labeled to generate cDNA probes. The probes are then hybridized to an array of nucleic acids immobilized on a solid support. The array is configured such that the sequence and position of each member of the array is known. For example, a selection of genes that have potential to be expressed in certain disease states may be arrayed on a solid support. Hybridization of a labeled probe with a particular array member indicates that the sample from which the probe was derived expresses that gene. Differential gene expression analysis of disease tissue can provide valuable information. Microarray technology utilizes nucleic acid hybridization techniques and computing technology to evaluate the mRNA expression profile of thousands of genes within a single experiment. (See, e.g., WO 01/75166 published Oct. 11, 2001; see also, for example, U.S. Pat. Nos. 5,700,637, 5,445,934, and 5,807,522, Lockart, Nature Biotechnology, 14:1675-1680 (1996); Cheung, V. G. et al., Nature Genetics 21(Suppl):15-19 (1999) for a discussion of array fabrication). DNA microarrays are miniature arrays containing gene fragments that are either synthesized directly onto or spotted onto glass or other substrates. Thousands of genes are usually represented in a single array. A typical microarray experiment involves the following steps: 1) preparation of fluorescently labeled target from RNA isolated from the sample, 2) hybridization of the labeled target to the microarray, 3) washing, staining, and scanning of the array, 4) analysis of the scanned image and 5) generation of gene expression profiles. Currently two main types of DNA microarrays are being used: oligonucleotide (usually 25 to 70 mers) arrays and gene expression arrays containing PCR products prepared from cDNAs. In forming an array, oligonucleotides can be either prefabricated and spotted to the surface or directly synthesized on to the surface (in situ).
The Affymetrix GeneChip® system is a commercially available microarray system which comprises arrays fabricated by direct synthesis of oligonucleotides on a glass surface. Probe/Gene Arrays: Oligonucleotides, usually 25 mers, are directly synthesized onto a glass wafer by a combination of semiconductor-based photolithography and solid phase chemical synthesis technologies. Each array contains up to 400,000 different oligos and each oligo is present in millions of copies. Since oligonucleotide probes are synthesized in known locations on the array, the hybridization patterns and signal intensities can be interpreted in terms of gene identity and relative expression levels by the Affymetrix Microarray Suite software. Each gene is represented on the array by a series of different oligonucleotide probes. Each probe pair consists of a perfect match oligonucleotide and a mismatch oligonucleotide. The perfect match probe has a sequence exactly complimentary to the particular gene and thus measures the expression of the gene. The mismatch probe differs from the perfect match probe by a single base substitution at the center base position, disturbing the binding of the target gene transcript. This helps to determine the background and nonspecific hybridization that contributes to the signal measured for the perfect match oligo. The Microarray Suite software subtracts the hybridization intensities of the mismatch probes from those of the perfect match probes to determine the absolute or specific intensity value for each probe set. Probes are chosen based on current information from GenBank and other nucleotide repositories. The sequences are believed to recognize unique regions of the 3′ end of the gene. A GeneChip Hybridization Oven (“rotisserie” oven) is used to carry out the hybridization of up to 64 arrays at one time. The fluidics station performs washing and staining of the probe arrays. It is completely automated and contains four modules, with each module holding one probe array. Each module is controlled independently through Microarray Suite software using preprogrammed fluidics protocols. The scanner is a confocal laser fluorescence scanner which measures fluorescence intensity emitted by the labeled cRNA bound to the probe arrays. The computer workstation with Microarray Suite software controls the fluidics station and the scanner. Microarray Suite software can control up to eight fluidics stations using preprogrammed hybridization, wash, and stain protocols for the probe array. The software also acquires and converts hybridization intensity data into a presence/absence call for each gene using appropriate algorithms. Finally, the software detects changes in gene expression between experiments by comparison analysis and formats the output into .txt files, which can be used with other software programs for further data analysis.
In some embodiments, expression of various biomarkers in a sample may also be assessed by examining gene deletion or gene amplification. Gene deletion or amplification may be measured by any one of a wide variety of protocols known in the art, for example, by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA (Thomas, Proc. Natl. Acad. Sci. USA, 77:5201-5205 (1980)), dot blotting (DNA analysis), or in situ hybridization (e.g., FISH), using an appropriately labeled probe, cytogenetic methods or comparative genomic hybridization (CGH) using an appropriately labeled probe. By way of example, these methods may be employed to detect deletion or amplification of genes listed in any of Tables 2-4, 6, 7 and 13.
In some embodiments, expression of various biomarkers in a sample may be assessed by hybridization assays using complementary DNA probes (such as in situ hybridization using labeled riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers, such as primers specific for one or more genes listed in any of Tables 2-4, 6, 7 and 13, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like).
Tissue or cell samples from mammals can be conveniently assayed for, e.g., mRNAs of genes listed in any of Tables 2-4, 6, 7 and 13, using Northern, dot blot or PCR analysis. In some embodiments, expression of one or more biomarkers may be assayed by RT-PCR. In some embodiments, the RT-PCR may be quantitative RT-PCR (qRT-PCR). In some embodiments, the RT-PCR is real-time RT-PCR. In some embodiments, the RT-PCR is quantitative real-time RT-PCR. RT-PCR assays such as quantitative PCR assays are well known in the art. In an illustrative embodiment of the invention, a method for detecting a mRNA in a biological sample comprises producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using a polynucleotide as sense and antisense primers to amplify cDNAs therein; and detecting the presence of the amplified cDNA of interest. In some embodiments, the real-time RT-PCR may be quantitative RT-PCR. In some embodiments, the real-time RT-PCR may be performed using TaqMan® chemistry (Applied Biosystems). In some embodiments, the real-time RT-PCR may be performed using TaqMan® chemistry (Applied Biosystems) and the ABI Prism® 7700 Sequence Detection System (Applied Biosystems). The real-time RT-PCR combines the principles that Taq polymerase has a 5′-3; exonuclease activity and dual-labeled fluorogenic oligonucleotide problems have been created which emit a fluorescent signal only upon cleavage, based on the principle of fluorescence resonance energy transfer. See, e.g., Overbergh, L. et al., J. Biomolecular Techniques 14(1): 33-43 (2003). In addition, such methods can include one or more steps that allow one to determine the levels of mRNA, such as a mRNA of genes listed in any of Tables 2-4, 6, 7 and 13, in a biological sample (e.g., by simultaneously examining the levels a comparative control mRNA sequence of a “housekeeping” gene such as an actin family member and/or one or more genes listed in Table 8 or Table 9). Examples of primers and probes that may be used for conducting qRT-PCR are provided in Table 10.
In some embodiments, the expression of proteins encoded by the genes listed in any of Tables 2-4, 6, 7 and 13 in a sample is examined using immunohistochemistry and staining protocols. Immunohistochemical staining of tissue sections has been shown to be a reliable method of assessing or detecting presence of proteins in a sample. Immunohistochemistry (“IHC”) techniques utilize an antibody to probe and visualize cellular antigens in situ, generally by chromogenic or fluorescent methods.
For sample preparation, a tissue or cell sample from a mammal (typically a human patient) may be used. Examples of samples include, but are not limited to, tissue biopsy, blood, lung aspirate, sputum, lymph fluid, etc. The sample can be obtained by a variety of procedures known in the art including, but not limited to surgical excision, aspiration or biopsy. The tissue may be fresh or frozen. In some embodiments, the sample is fixed and embedded in paraffin or the like.
The tissue sample may be fixed (i.e. preserved) by conventional methodology (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology,” 3rd edition (1960) Lee G. Luna, H T (ASCP) Editor, The Blakston Division McGraw-Hill Book Company, New York; The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology (1994) Ulreka V. Mikel, Editor, Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C.). One of skill in the art will appreciate that the choice of a fixative is determined by the purpose for which the sample is to be histologically stained or otherwise analyzed. One of skill in the art will also appreciate that the length of fixation depends upon the size of the tissue sample and the fixative used. By way of example, neutral buffered formalin, Bouin's or paraformaldehyde, may be used to fix a sample.
Generally, the sample is first fixed and is then dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned. Alternatively, one may section the tissue and fix the sections obtained. By way of example, the tissue sample may be embedded and processed in paraffin by conventional methodology (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology”, supra). Examples of paraffin that may be used include, but are not limited to, Paraplast, Broloid, and Tissuemay. Once the tissue sample is embedded, the sample may be sectioned by a microtome or the like (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology”, supra). By way of example for this procedure, sections may range from about three microns to about five microns in thickness. Once sectioned, the sections may be attached to slides by several standard methods. Examples of slide adhesives include, but are not limited to, silane, gelatin, poly-L-lysine and the like. By way of example, the paraffin embedded sections may be attached to positively charged slides and/or slides coated with poly-L-lysine.
If paraffin has been used as the embedding material, the tissue sections are generally deparaffinized and rehydrated to water. The tissue sections may be deparaffinized by several conventional standard methodologies. For example, xylenes and a gradually descending series of alcohols may be used (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology”, supra). Alternatively, commercially available deparaffinizing non-organic agents such as Hemo-De7 (CMS, Houston, Tex.) may be used.
In some embodiments, subsequent to the sample preparation, a tissue section may be analyzed using IHC. IHC may be performed in combination with additional techniques such as morphological staining and/or fluorescence in-situ hybridization. Two general methods of IHC are available; direct and indirect assays. According to the first assay, binding of antibody to the target antigen (e.g., a protein or fragment thereof encoded by one or more genes listed in Tables 1-4, 6 and 7) is determined directly. This direct assay uses a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction. In a typical indirect assay, unconjugated primary antibody binds to the antigen and then a labeled secondary antibody binds to the primary antibody. Where the secondary antibody is conjugated to an enzymatic label, a chromogenic or fluorogenic substrate is added to provide visualization of the antigen. Signal amplification occurs because several secondary antibodies may react with different epitopes on the primary antibody.
The primary and/or secondary antibody used for immunohistochemistry typically will be labeled with a detectable moiety. Numerous labels are available which can be generally grouped into the following categories:
(a) Radioisotopes, such as 35S, 14C, 125I, 3H, and 131I. The antibody can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
(b) Colloidal gold particles.
(c) Fluorescent labels including, but are not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE7 and SPECTRUM GREEN7 and/or derivatives of any one or more of the above. The fluorescent labels can be conjugated to the antibody using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter.
(d) Various enzyme-substrate labels are available and U.S. Pat. No. 4,275,149 provides a review of some of these. The enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above. The chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor. Examples of enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, β-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like. Techniques for conjugating enzymes to antibodies are described in O'Sullivan et al., Methods for the Preparation of Enzyme-Antibody Conjugates for use in Enzyme Immunoassay, in Methods in Enzym. (ed. J. Langone & H. Van Vunakis), Academic press, New York, 73:147-166 (1981).
Examples of enzyme-substrate combinations include, for example:
(i) Horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor (e.g., orthophenylene diamine (OPD) or 3,3′,5,5′-tetramethyl benzidine hydrochloride (TMB));
(ii) alkaline phosphatase (AP) with para-Nitrophenyl phosphate as chromogenic substrate; and
(iii) β-D-galactosidase (β-D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl-1′-D-galactosidase) or fluorogenic substrate (e.g., 4-methylumbelliferyl-β-D-galactosidase).
Numerous other enzyme-substrate combinations are available to those skilled in the art. For a general review of these, see U.S. Pat. Nos. 4,275,149 and 4,318,980. Sometimes, the label is indirectly conjugated with the antibody. The skilled artisan will be aware of various techniques for achieving this. For example, the antibody can be conjugated with biotin and any of the four broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner. Alternatively, to achieve indirect conjugation of the label with the antibody, the antibody is conjugated with a small hapten and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody. Thus, indirect conjugation of the label with the antibody can be achieved.
Aside from the sample preparation procedures discussed above, further treatment of the tissue section prior to, during or following IHC may be desired. For example, epitope retrieval methods, such as heating the tissue sample in citrate buffer may be carried out (see, e.g., Leong et al. Appl. Immunohistochem. 4(3):201 (1996)).
Following an optional blocking step, the tissue section is exposed to primary antibody for a sufficient period of time and under suitable conditions such that the primary antibody binds to the target protein antigen in the tissue sample. Appropriate conditions for achieving this can be determined by routine experimentation. The extent of binding of antibody to the sample is determined by using any one of the detectable labels discussed above. Preferably, the label is an enzymatic label (e.g. HRPO) which catalyzes a chemical alteration of the chromogenic substrate such as 3,3′-diaminobenzidine chromogen. Preferably the enzymatic label is conjugated to antibody which binds specifically to the primary antibody (e.g. the primary antibody is rabbit polyclonal antibody and secondary antibody is goat anti-rabbit antibody).
In some embodiments, the antibodies employed in the IHC analysis to detect expression of one or more biomarkers are antibodies generated to bind primarily to the one or more biomarkers of interest, such as one or more proteins encoded by genes listed in any of Tables 2-4, 6 and 7. In some embodiments, the antibody is a monoclonal antibody. Antibodies are readily available in the art, including from various commercial sources, and can also be generated using routine skills known in the art.
Specimens thus prepared may be mounted and coverslipped. Slide evaluation is then determined, e.g. using a microscope, and staining intensity criteria, routinely used in the art, may be employed. As one example, staining intensity criteria may be evaluated as follows:
TABLE A
Staining Pattern Score
No staining is observed in cells. 0
Faint/barely perceptible staining is detected in 1+
more than 10% of the cells.
Weak to moderate staining is observed in 2+
more than 10% of the cells.
Moderate to strong staining is observed in 3+
more than 10% of the cells.
In alternative methods, the sample may be contacted with an antibody specific for said biomarker under conditions sufficient for an antibody-biomarker complex to form, and then detecting said complex. The presence of the biomarker may be detected in a number of ways, such as by Western blotting and ELISA procedures for assaying a wide variety of tissues and samples, including plasma or serum. A wide range of immunoassay techniques using such an assay format are available, see, e.g., U.S. Pat. Nos. 4,016,043, 4,424,279 and 4,018,653. These include both single-site and two-site or “sandwich” assays of the non-competitive types, as well as in the traditional competitive binding assays. These assays also include direct binding of a labeled antibody to a target biomarker.
Sandwich assays are among the most useful and commonly used assays. A number of variations of the sandwich assay technique exist, and all are intended to be encompassed by the present invention. Briefly, in a typical forward assay, an unlabelled antibody is immobilized on a solid substrate, and the sample to be tested brought into contact with the bound molecule. After a suitable period of incubation, for a period of time sufficient to allow formation of an antibody-antigen complex, a second antibody specific to the antigen, labeled with a reporter molecule capable of producing a detectable signal is then added and incubated, allowing time sufficient for the formation of another complex of antibody-antigen-labeled antibody. Any unreacted material is washed away, and the presence of the antigen is determined by observation of a signal produced by the reporter molecule. The results may either be qualitative, by simple observation of the visible signal, or may be quantitated by comparing with a control sample containing known amounts of biomarker.
Variations on the forward assay include a simultaneous assay, in which both sample and labeled antibody are added simultaneously to the bound antibody. These techniques are well known to those skilled in the art, including any minor variations as will be readily apparent. In a typical forward sandwich assay, a first antibody having specificity for the biomarker is either covalently or passively bound to a solid surface. The solid surface is typically glass or a polymer, the most commonly used polymers being cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene. The solid supports may be in the form of tubes, beads, discs of microplates, or any other surface suitable for conducting an immunoassay. The binding processes are well-known in the art and generally consist of cross-linking covalently binding or physically adsorbing, the polymer-antibody complex is washed in preparation for the test sample. An aliquot of the sample to be tested is then added to the solid phase complex and incubated for a period of time sufficient (e.g., 2-40 minutes or overnight if more convenient) and under suitable conditions (e.g., from room temperature to 40° C. such as between 25° C. and 32° C. inclusive) to allow binding of any subunit present in the antibody. Following the incubation period, the antibody subunit solid phase is washed and dried and incubated with a second antibody specific for a portion of the biomarker. The second antibody is linked to a reporter molecule which is used to indicate the binding of the second antibody to the molecular marker.
In some embodiments, the methods involves immobilizing the target biomarkers in the sample and then exposing the immobilized target to specific antibody which may or may not be labeled with a reporter molecule. Depending on the amount of target and the strength of the reporter molecule signal, a bound target may be detectable by direct labeling with the antibody. Alternatively, a second labeled antibody, specific to the first antibody is exposed to the target-first antibody complex to form a target-first antibody-second antibody tertiary complex. The complex is detected by the signal emitted by the reporter molecule. By “reporter molecule”, as used in the present specification, is meant a molecule which, by its chemical nature, provides an analytically identifiable signal which allows the detection of antigen-bound antibody. The most commonly used reporter molecules in this type of assay are either enzymes, fluorophores or radionuclide containing molecules (i.e. radioisotopes) and chemiluminescent molecules.
In the case of an enzyme immunoassay, an enzyme is conjugated to the second antibody, generally by means of glutaraldehyde or periodate. As will be readily recognized, however, a wide variety of different conjugation techniques exist, which are readily available to the skilled artisan. Commonly used enzymes include horseradish peroxidase, glucose oxidase, -galactosidase and alkaline phosphatase, amongst others. The substrates to be used with the specific enzymes are generally chosen for the production, upon hydrolysis by the corresponding enzyme, of a detectable color change. Examples of suitable enzymes include alkaline phosphatase and peroxidase. It is also possible to employ fluorogenic substrates, which yield a fluorescent product rather than the chromogenic substrates noted above. In all cases, the enzyme-labeled antibody is added to the first antibody-molecular marker complex, allowed to bind, and then the excess reagent is washed away. A solution containing the appropriate substrate is then added to the complex of antibody-antigen-antibody. The substrate will react with the enzyme linked to the second antibody, giving a qualitative visual signal, which may be further quantitated, usually spectrophotometrically, to give an indication of the amount of biomarker which was present in the sample. Alternately, fluorescent compounds, such as fluorescein and rhodamine, may be chemically coupled to antibodies without altering their binding capacity. When activated by illumination with light of a particular wavelength, the fluorochrome-labeled antibody adsorbs the light energy, inducing a state to excitability in the molecule, followed by emission of the light at a characteristic color visually detectable with a light microscope. As in the EIA, the fluorescent labeled antibody is allowed to bind to the first antibody-molecular marker complex. After washing off the unbound reagent, the remaining tertiary complex is then exposed to the light of the appropriate wavelength, the fluorescence observed indicates the presence of the molecular marker of interest. Immunofluorescence and EIA techniques are both very well established in the art. However, other reporter molecules, such as radioisotope, chemiluminescent or bioluminescent molecules, may also be employed.
In some embodiments, expression of a selected biomarker in a tissue or cell sample may be examined by way of functional or activity-based assays. For instance, if the biomarker is an enzyme, one may conduct assays known in the art to determine or detect the presence of the given enzymatic activity in the tissue or cell sample.
In any of the above methods of assessing level of expression of one or more biomarkers, a sample comprising a target molecule can be obtained by methods well known in the art, and that are appropriate for the particular type and location of the disease of interest. Tissue biopsy is often used to obtain a representative piece of disease tissue. Alternatively, cells can be obtained indirectly in the form of tissues/fluids that are known or thought to contain the disease cells of interest. For instance, samples of disease lesions may be obtained by resection, bronchoscopy, fine needle aspiration, bronchial brushings, or from sputum, pleural fluid or blood. Genes or gene products can be detected from disease tissue or from other body samples such as urine, sputum or serum. The same techniques discussed above for detection of target genes or gene products in disease samples can be applied to other body samples. By screening such body samples, a simple early diagnosis can be achieved for these diseases. In addition, the progress of therapy can be monitored more easily by testing such body samples for target genes or gene products.
Means for enriching a tissue preparation for disease cells are known in the art. For example, the tissue may be isolated from paraffin or cryostat sections. Cells of interest may also be separated from normal cells by flow cytometry or laser capture microdissection. These, as well as other techniques for separating disease from normal cells, are well known in the art. If the disease tissue is highly contaminated with normal cells, detection of signature gene expression profile may be more difficult, although techniques for minimizing contamination and/or false positive/negative results are known, some of which are described herein below. For example, a sample may also be assessed for the presence of a biomarker (including a mutation) known to be associated with a disease cell of interest but not a corresponding normal cell, or vice versa.
Subsequent to the determination that the tissue or cell sample expresses one or more of the biomarkers indicating the tissue or cell sample will be sensitive to treatment with anti-CD40 antibodies, it is contemplated that an effective amount of the anti-CD40 antibody may be administered to the mammal, such as a human to treat a disorder, such as a B-cell lymphoma which is afflicting the mammal. Diagnosis in mammals, such as humans, of the various pathological conditions described herein can be made by the skilled practitioner.
Comparing Expression Levels and Predicting, Assessing or Aiding Assessment of Responsiveness of B-Cell Lymphoma to an Anti-CD40 Antibody Treatment
The methods described herein comprise a process of comparing a measured expression level of a marker gene and a reference level. The reference level may be a measured expression level of a reference gene different from the marker gene or a measured expression level of the same marker gene in a different sample.
In some embodiments, a measured expression level of a marker gene in a B cell lymphoma sample from a subject is compared to a measured expression level of a reference gene in the sample. In some embodiments, the expression level of the reference gene does not substantially change among various types of B lymphoma cells, including anti-CD40 antibody sensitive and resistant cells (e.g., genes in Table 8 or Table 9). In some embodiments, the ratio of the measured expression level of the marker gene to the measured expression level of the reference is calculated, and the ratio may be used for assessing or aiding assessment of responsiveness of the B cell lymphoma to an anti-CD antibody treatment.
In some embodiments, a measured expression level of a marker gene in a B cell lymphoma sample from a subject is compared to a measured expression level of the marker gene in a reference sample. In some embodiments, the reference sample comprises B lymphoma cells that are resistant or not responsive to an anti-CD40 antibody. For example, the comparison is performed to determine the magnitude of the difference between the measured expression levels of the marker gene in the sample from the subject and in the reference sample (e.g., comparing the fold or percentage difference between the expression levels of the marker gene in the sample from the subject and the reference sample). An increase or decreased expression of a marker gene in the sample from the subject as compared to the expression of the marker gene in the reference sample comprising B lymphoma cells that are resistant or not responsive to an anti-CD40 antibody suggests or indicates responsiveness of the B-cell lymphoma to treatment with an anti-CD40 antibody. See Table 4 for marker genes having increased and decreased expression in anti-CD40 antibody sensitive cells as compared to resistant cells. For examples, VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, and MEF2B are generally overexpressed in anti-CD40 antibody sensitive cells as compared to resistant cells. In some embodiments, a fold of increase in the expression level of the sample from the subject can be at least about any of 1.5×, 1.75×, 2×, 3×, 4×, 5×, 6×, 7×, 8×, 9×, or 10× the expression level of the reference sample. In some embodiments, a fold of decrease in the expression level of the sample from the subject can be less than about any of 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8 of the expression level of the reference sample.
In some embodiments, expression level of one or more marker genes selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are compared to a reference level.
In some embodiments, an increased expression level of one or more of IFITM1, CD79B, IGF1R, CD44, CTSC, EPDR1, and PUS7 as compared to a reference level indicates that said subject is less likely to respond to an agonist anti-CD40 antibody treatment. In some embodiments, the reference level is a value or a range determined by expression levels of the corresponding marker gene in samples comprising B lymphoma cells from subjects having tumor volume increased after an agonist anti-CD40 antibody treatment.
In some embodiments, an increased expression of one or more of CD40, RGS13, VNN2, LMO2, CD22, BTG2, and UAP1 as compared to a reference level indicates that said subject is likely to respond to the agonist anti-CD40 antibody treatment. In some embodiments, the reference level is a value or a range determined by expression levels of the corresponding marker gene in samples comprising B lymphoma cells from subjects having tumor volume decreased after an agonist anti-CD40 antibody treatment.
In some embodiments, the expression level BCL6 is measured and compared to a reference level. The expression level of BCL6 is used for predicting, assessing, or aiding assessment of responsiveness of the subject to an anti-CD40 antibody treatment. As shown in Example 2, BCL6 expression trends lower in those subjects with tumor increases after an agonist anti-CD40 antibody treatment. In some embodiments, an increased expression of BCL6 as compared to a reference level determined by expression level of BCL6 in samples from subjects having tumor volume decreased after an agonist anti-CD40 antibody treatment may indicate the subject is likely to respond to the agonist anti-CD40 antibody treatment.
In some embodiments, the expression levels of marker genes in Table 7) are measured, and a sensitivity index calculated as the sum of signed t-scores for log 2-scale expression of genes pairs 1-8 in Table 7 is determined, wherein a sensitivity index greater than −4 suggests or indicates the B-cell lymphoma is responsive to an anti-CD40 antibody treatment. In some embodiments, the sensitivity index is greater than −3, greater than −2, greater than −1, or greater than 0. In some embodiments, the sensitivity index is between −4 and 20. In some embodiments, the sensitivity index is between 0 and 20.
In some embodiments, the expression levels of one or more of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, UAP1, and PUS7 are measured, and a sensitivity index is calculated based on the measured expression level of the marker genes. For example, the following equation may be used for determining sensitivity index (SI):
SI = j = 1 p β j x j - μ ^ j σ ^ j 2
wherein expression level of at least one marker gene having a positive correlation value and at least one marker gene having a negative correlation value shown in Table 13 are measured; wherein (i) βj is the coefficient value for each marker genes measured; (ii) p is the number of marker genes measured; (iii) xi is transformed, normalized expression level for the sample from the subject for expression level of each marker measured; and (iv) μj and σj are means and standard deviations for each marker gene measured; wherein βj, μj and σj are determined from patient samples comprising B lymphoma cells from a clinical trial. In some embodiments, a value equals or greater than zero for the sensitivity index indicates that the subject is likely to respond the anti-CD40 antibody treatment, or wherein a value less than zero for the sensitivity index indicates that the subject is less likely to respond the anti-CD40 antibody treatment. Example 2 described in detail how to analyze and determine parameters for reference samples and new samples. In some embodiments, the expression level of IFITM1, RGS13, CD79B, CD22, BTG2, CD44, EPDR1, and UAP1 are measured and used for the sensitivity index calculation. In some embodiments, equal number of positive correlated marker genes and negative correlated marker genes are measured and used for the sensitivity index calculation.
Methods for determining sensitivity index are known in the art. See Zhou H. and Hastie T. (2005) Regularization and variable selection via the elastic net; J. R. Statist. Soc. B. 67(2). pp. 301-320; Friedman J., Hastie T. and Tibshirani R. 2008. Regularization Paths for Generalized Linear Models via Coordinate Descent. Technical Report, Department of Statistics, Stanford University (World Wide Web-stat.stanford.edu/˜hastie/Papers/glmnet.pdf) R package glmnet; R Development Core Team (2008). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. ISBN 3-900051-07-0, URL World Wide Web at R-project.org.
The comparison can be carried out in any convenient manner appropriate to the type of measured value and reference value for the gene markers at issue. The process of comparing may be manual or it may be automatic. In some embodiments, measured expression levels are normalized values. For example, the expression level may be normalized based on the equation under Transformed, Normalized Assay Values described in Example 2. As will be apparent to those of skill in the art, replicate measurements may be taken for the expression levels of marker genes and/or reference genes. In some embodiments, replicate measurements are taking into account for the measured values. The replicate measurements may be taken into account by using either the mean or median of the measured values as the “measured value”. Statistical analysis known in the art may be used to verify the significance of the difference between the two values compared.
Anti-CD40 Antibody Treatment
The marker genes identified in the invention may be used for predicting, assessing, or aiding assessment of responsiveness of B-cell lymphoma to treatment with one or more anti-CD40 antibodies. The anti-CD40 antibodies may be one or more agonist antibodies (i.e., bind and stimulate CD40). Stimulatory antibodies can be of different types, such as: (1) those that deliver a stimulatory signal through CD40 but do not increase the interaction between CD40 and CD40L (e.g., antibody G28-5 and antibodies derived from G28-5 described in U.S. Pat. No. 5,182,368; and PCT WO 96/18413), or decrease the interaction between CD40 and CD40L (e.g., antibodies HuCD40-M2 and HuCD40-M3 and humanized antibodies described in U.S. Pat. No. 5,674,492; and (2) those that deliver a stimulatory signal through CD40 and can increase the interaction between CD40 and CD40L, e.g., S2C6 (Francisco et al., 2000, Cancer Res. 60:3225-31) and antibodies derived from S2C6. Agonists antibodies are also described in U.S. Pat. No. 7,288,251. The anti-CD40 antibodies may be one or more antagonist antibodies (i.e., bind CD40 and inhibit activities induced by CD40L). Examples of antagonist anti-CD40 antibodies include human antibody CHIR-12.12 described in U.S. Pub. No. 2007/0110754, and anti-CD40 antibodies described in WO 97/31025.
The methods of the invention may further comprise administering an effective amount of an anti-CD40 antibody to a subject having a B-cell lymphoma after the subject has been identified as a candidate for treatment based on the assays/methods described herein. One or more anti-CD40 antibodies may be administered. In some embodiments, the anti-CD40 antibody is administered in conjunction with one or more of the following therapeutic agents: rituximab, gemzar, and ICE. For example, an anti-CD40 antibody can be administered to the patient in conjunction with rituximab therapy; with rituximal plus gemzar; with rituximal plus ICE (ifosfamide, carboplatin, etoposide) (R-ICE); or with rituximab plus chemotherapy.
As used herein, administration “in conjunction” includes simultaneous administration and/or administration at different times. Administration in conjunction also encompasses administration as a co-formulation (i.e., different drugs are present in the same composition) or administration as separate compositions, administration at different dosing frequencies or intervals, and administration using the same route or different routes.
The anti-CD40 antibodies or functional fragments can be used for the treatment of patients with NHL that are nonresponsive or have an inadequate response to treatment with any one of the following drugs: rituximab (Genentech); ocrelizumab (Genentech, Inc.); ibritumomab tiuxetan (Zevalin™, Biogen Idec); tositumomab (Bexxar™, GlaxoSmithKline); HuMAX-CD20™ (GenMab); IMMU-106 (which is a humanized anti-CD20 a.k.a. hA20 or 90Y-hLL2, Immunomedics); AME-133 (Applied Molecular Evolution/Eli Lilly); gentuzumab ozogamicin (Mylotarg™, a humanized anti-CD33 antibody, Wyeth/PDL); alemtuzumab (Campath™, an anti-CD52 antibody, Schering Plough/Genzyme); epratuzumab (IMMU-103™, a humanized anti-CD22 antibody, Immunomedics), or have relapsed after treatment with these drugs.
The following references describe lymphomas and CLL, their diagnoses, treatment and standard medical procedures for measuring treatment efficacy. Canellos G P, Lister, T A, Sklar J L: The Lymphomas. W.B.Saunders Company, Philadelphia, 1998; van Besien K and Cabanillas, F: Clinical Manifestations, Staging and Treatment of Non-Hodgkin's Lymphoma, Chap. 70, pp 1293-1338, in: Hematology, Basic Principles and Practice, 3rd ed. Hoffman et al. (editors). Churchill Livingstone, Philadelphia, 2000; and Rai, K and Patel, D: Chronic Lymphocytic Leukemia, Chap. 72, pp 1350-1362, in: Hematology, Basic Principles and Practice, 3rd ed. Hoffman et al. (editors). Churchill Livingstone, Philadelphia, 2000.
Anti-CD40 antibodies for use in the treatment include chimeric, humanized and human antibodies. Any agonist or antagonist antibodies described herein or known in the art may be used in the treatment. For example, humanized anti-CD40 antibodies described in WO 2006/128103 may be used for the anti-CD40 antibody treatment, and these antibodies and their amino acid sequences are incorporated herein by reference. In some embodiments, the anti-CD40 antibody for used in the treatment described herein binds to CD40 (such as human CD40) expressed on B lymphoma cells and induces apoptosis of the B lymphoma cells. The anti-CD40 antibody may also have the characteristics of killing B lymphoma cells in vivo via immune effector functions, such as ADCC, CDC, and/or ADCP. In some embodiments, the anti-CD40 antibody binds to CD40 with a Kd value of no higher than about 1×10−8 or no higher than 1×10−9. In some embodiments, the anti-CD40 antibody binds to CD40 and stimulates CD40 (i.e., an agonist antibody). In some embodiments, the anti-CD40 antibody increases the binding of CD40 ligand to CD40, for example, by at least 45%, by at least 50%, by at least 60%, or by at least 75%. A method of determining increases in binding of CD40 ligand to CD40 are disclosed in U.S. Pat. No. 6,838,261 (the disclosure of which is incorporated by reference herein). In some embodiments, the anti-CD40 is a humanized antibody derived from murine monoclonal antibody S2C6 described in WO 00/75348 (including antibodies provided in Tables 3 and 4 of WO 00/75348). In some embodiments, the anti-CD40 antibody comprises the heavy chain amino acid sequence shown in SEQ ID NO:1 and the light chain amino acid sequence shown in SEQ ID NO:2, for example anti-CD40 Ab.1.
D. Kits
For use in the applications described or suggested above, kits or articles of manufacture are also provided by the invention. Such kits may comprise at least one reagent specific for detecting expression level of a marker gene described herein, and may further include instructions for carrying out a method described herein.
In some embodiments, the invention provides compositions and kits comprising primers and primer pairs, which allow the specific amplification of the polynucleotides of the invention or of any specific parts thereof, and probes that selectively or specifically hybridize to nucleic acid molecules of the invention or to any part thereof. Probes may be labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme. Such probes and primers can be used to detect the presence of polynucleotides, such as the polynucleotides corresponding to genes listed in Table 1-4, 6, 7 and 13, in a sample and as a means for detecting a cell expressing proteins encoded by the polynucleotides corresponding to genes listed in Table 1-4, 6, 7 and 13. As will be understood by the skilled artisan, a great many different primers and probes may be prepared based on the sequences provided in herein and used effectively to amplify, clone and/or determine the presence and/or levels of mRNAs.
In some embodiments, the kits comprise reagents for detecting expression levels of at least two, at least three, at least five, at least ten, at least fifteen, at least twenty marker genes. Kits may also comprise reference samples that are useful as generating reference values. The marker genes include, but are not limited to VNN2, MEF2C, LTB, KCNN3, NCF1, BCL6, IGJ, ELTI1902, PNOC, CSF2RB, POU2AF1, CD22, RGS13, MEF2B, LRRC8A, CD40, IFITM1, SMN1, PRRCA, EPDR1, PRPSAP2, IGF1R, BTG2, LMO2, YIPF3, CD79B, CD44, CTSC, UAP1, and PUS7. The reagents for detecting mRNA expression level of a marker gene may comprise at least one pair of primers specific for amplifying the mRNA products of one marker gene. In some embodiments, the pair of primers may target the 3′ end of the mRNA sequence (e.g., targeting mRNA at the 3′ UTR which is usually shared in common with all transcript variants). In some embodiments, the kits may further comprise a surface or substrate (such as a microarray) for capture probes for detecting of amplified nucleic acids.
In some embodiments, the kits comprises at least one pair of primers and a probe specific for detecting one marker gene expression level using qRT-PCR. Examples of sets of primers and probes that can be used in qRT-PCR are shown in Table 10. For detecting IFITM1, primer and probe sets shown in SEQ ID NOS:27, 28 and 29, SEQ ID NOS:60, 61, and 62, and SEQ ID NOS:93, 94, and 95 may be used. For detecting CD40, primer and probe sets shown in SEQ ID NOS:24, 25, and 26, SEQ ID NOS:57, 58, and 59, SEQ ID NOS:90, 91 and 92 may be used. For detecting RGS13, primer and probe sets shown in SEQ ID NOS:114, 115, and 116, and SEQ ID NOS:126, 127, and 128 may be used. For detecting VNN2, primer and probe sets shown in SEQ ID NOS:30, 31, and 32, SEQ ID NOS:63, 64, and 65, and SEQ ID NOS:96, 97, and 98. For detecting LMO2, primer and probe sets shown in SEQ ID NOS:12, 13, and 14, SEQ ID NOS:45, 46, and 47, and SEQ ID NOS:78, 79, and 80. For detecting CD79B, primer and probe sets shown in SEQ ID NOS:141, 142, and 143, SEQ ID NOS:150, 151, and 152, and SEQ ID NOS:159, 160, and 161. For detecting CD22, primer and probe sets shown in SEQ ID NOS:15, 16, and 17, SEQ ID NOS:48, 49, and 50, and SEQ ID NOS:81, 82, and 83. For detecting BTG2, primer and probe sets shown in SEQ ID NOS:9, 10, and 11, SEQ ID NOS:42, 43, and 44, and SEQ ID NOS:75, 76, and 77. For detecting IGF1R, primer and probe sets shown in SEQ ID NOS:6, 7, and 8, SEQ ID NOS:39, 40, and 41, and SEQ ID NOS:72, 73, and 74. For detecting CD44, primer and probe sets shown in SEQ ID NOS:174, 175, and 176, SEQ ID NOS:180, 181, and 182, and SEQ ID NOS:186, 187, and 188. For detecting CTSC, primer and probe sets shown in SEQ ID NOS:165, 166, and 167, SEQ ID NOS:168, 169, and 170, and SEQ ID NOS:171, 172, and 173. For detecting EPDR1, primer and probe sets shown in SEQ ID NOS:21, 22, and 23, SEQ ID NOS:54, 55, and 56, SEQ ID NOS:87, 88, and 89, SEQ ID NOS:129, 130, and 131, SEQ ID NOS:132, 133, and 134, SEQ ID NOS:135, 136, and 137. For detecting UAP1, primer and probe sets shown in SEQ ID NOS:138, 139, and 140, SEQ ID NOS:147, 148, and 149, and SEQ ID NOS:156, 157, and 158. For detecting PUS7, primer and probe sets shown in SEQ ID NOS:177, 178, and 179, SEQ ID NOS:183, 184, and 185, and SEQ ID NOS:189, 190, and 191. For detecting BCL6, primer and probe sets shown in SEQ ID NOS:102, 103, and 104, and SEQ ID NOS:108, 109, and 110.
The reagents for detecting protein expression level of a marker gene may comprise an antibody that specifically binds to the protein encoded by the marker gene.
The kits may further comprise a carrier means being compartmentalized to receive in close confinement one or more container means such as vials, tubes, and the like, each of the container means comprising one of the separate elements to be used in the method. For example, one of the container means may comprise a probe that is or can be detectably labeled. Such probe may be an antibody or polynucleotide specific for a marker gene. Where the kit utilizes nucleic acid hybridization to detect the target nucleic acid, the kit may also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence and/or a container comprising a reporter-means, such as a biotin-binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, florescent, or radioisotope label.
The kit of the invention will typically comprise the container described above and one or more other containers comprising materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. A label may be present on the container to indicate that the composition is used for a specific therapy or non-therapeutic application, and may also indicate directions for either in vivo or in vitro use, such as those described above.
The kit can further comprise a set of instructions and materials for preparing a tissue or cell sample and preparing nucleic acid (such as mRNA) from the sample.
The invention provides a variety of compositions suitable for use in performing methods of the invention, which may be used in kits. For example, the invention provides surfaces, such as arrays that can be used in such methods. In some embodiments, an array of the invention comprises individual or collections of nucleic acid molecules useful for detecting mutations of the invention. For instance, an array of the invention may comprises a series of discretely placed individual nucleic acid oligonucleotides or sets of nucleic acid oligonucleotide combinations that are hybridizable to a sample comprising target nucleic acids, whereby such hybridization is indicative of presence or absence of a mutation of the invention.
Several techniques are well-known in the art for attaching nucleic acids to a solid substrate such as a glass slide. One method is to incorporate modified bases or analogs that contain a moiety that is capable of attachment to a solid substrate, such as an amine group, a derivative of an amine group or another group with a positive charge, into nucleic acid molecules that are synthesized. The synthesized product is then contacted with a solid substrate, such as a glass slide, which is coated with an aldehyde or another reactive group which will form a covalent link with the reactive group that is on the amplified product and become covalently attached to the glass slide. Other methods, such as those using amino propryl silican surface chemistry are also known in the art, as disclosed at world wide web at cmt.corning.com and cmgm.stanford.edu/pbrown1.
Attachment of groups to oligonucleotides which could be later converted to reactive groups is also possible using methods known in the art. Any attachment to nucleotides of oligonucleotides will become part of oligonucleotide, which could then be attached to the solid surface of the microarray. Amplified nucleic acids can be further modified, such as through cleavage into fragments or by attachment of detectable labels, prior to or following attachment to the solid substrate, as required and/or permitted by the techniques used.
The following are examples of the methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.
EXAMPLES Example 1 Identification of Predictive Genetic Markers for Responsiveness of NHL Patients to Anti-CD40 Antibody Treatment
Materials and Methods
Cell Viability Assays
NHL Cells were seeded in 384 well plates at 1500-5000 cells/well in 50 ul RPMI 1640 supplemented with 2% FBS and treated with serial concentrations of crosslinked anti-CD40 Ab.1 or control antibody (anti-gD 5B6). For crosslinking, anti-CD40 Ab.1 or anti-gD was incubated with F(ab′)2 fragments of a goat anti human IgG Fcγ fragment-specific antibody (Jackson ImmunoResearch, West Grove, Pa.) in a 1:4 ratio in medium for 30 minutes at room temperature before adding to cells. After 96 hours of incubation, cell viability was evaluated using CellTiter-Glo Luminescent Cell Viability Assay (Promega, Madison, Wis.) according to the manufacturer's instructions. Each data point was performed in quadruplicate.
XLfit was used to calculate IC50, IC25 and maximum inhibition. Data are expressed as average of three independent experiments. Sensitivity to anti-CD40 Ab.1 was binned into three categories: Sensitive, Intermediate, and Resistant based on IC25 and IC50 values.
Antibody
anti-CD40 Ab.1 is a humanized IgG1 mAb against CD40. It is produced in and secreted by a genetically engineered Chinese Hamster Ovary (CHO) cell line. The anti-CD40 Ab.1 used in the examples and referred to as anti-CD40 Ab.1 has the following amino acid sequence:
Heavy Chain (SEQ ID NO:1). The italicized underlined ASN 294 residue identifies the location of the carbohydrate moiety.
EVQLVESGGG LVQPGGSLRL SCAASGYSFT GYYIHWVRQA PGKGLEWVAR 50
VIPNAGGTSY NQKFKGRFTL SVDNSKNTAY LQMNSLRAED TAVYYCAREG 100
IYWWGQGTLV TVSSASTKGP SVFPLAPSSK STSGGTAALG CLVKDYFPEP 150
VTVSWNSGAL TSGVHTFPAV LQSSGLYSLS SVVTVPSSSL GTQTYICNVN 200
HKPSNTKVDK KVEPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI 250
SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV 300
SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP 350
SREEMTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS 400
FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL SPG 443
Light Chain (SEQ ID NO:2).
DIQMTQSPSS LSASVGDRVT ITCRSSQSLV HSNGNTFLHW YQQKPGKAPK 50
LLIYTVSNRF SGVPSRFSGS GSGTDFTLTI SSLQPEDFAT YFCSQTTHVP 100
WTFGQGTKVE IKRTVAAPSV FIFPPSDEQL KSGTASVVCL LNNFYPREAK 150
VQWKVDNALQ SGNSQESVTE QDSKDSTYSL SSTLTLSKAD YEKHKVYACE 200
VTHQGLSSPV TKSFNRGEC 219
Generation and Analysis of Gene Expression Profiles
Total RNA was extracted with the mirVana™ miRNA Isolation Kit (Ambion, Austin, Tex.) and was assayed using Affymetrix HGU133P2 whole genome expression microarrays. Raw data was extracted using an Affymetrix scanner and the resulting CEL files were processed using gcRMA with defaults in R Bioconductor Package (world wide web at bioconductor.org). Significantly differentially expressed genes were identified using a moderated t-test for differences across anti-CD40 Ab.1 sensitivity and viability classes. Further parameters were assessed using the LIMA package and t-statistics, p-values, adjusted p-values, and B-statistics were calculated for each gene. Probes were mapped to each gene and a 1:1 probe to gene mapping was selected for downstream analysis using the probe most strongly associated with the measure of sensitivity. For classification into Sensitive or Intermediate versus Resistant groups, quantitative stepwise linear modeling was combined with qualitative analysis of target pathways to identify a parsimonious set of genes to inclusion in the assay. Further details and results are provided in the Results (Table 7).
Gene set enrichment analysis was determined by utilizing the GSEA module within Gene Pattern (world wide web at genepattern.org). The enrichment score awards pre-specified classes of genes when their members are significantly differentially expressed in a concordant manner across phenotypes. The normalized enrichment score is calculated by taking the enrichment score and adjusting for the number of genes within a gene set. The nominal p-value is determined by permutating the sensitive and resistant labels and recomputing the normalized enrichment score to give a null distribution.
Anti-CD40 Ab.1 Sensitivity Index Identified Using Stepwise Linear Modeling
Each Target Gene is shown with its corresponding inversely correlated (anti-correlated) Pair Gene (Table 7), in order of the step at which the Target Gene was chosen for inclusion in the Index. The first 3 Main Genes (VNN2, RGS13, CD22 in Table 7) were selected from Tables 2-4 (Step 1) to model the dominant component of differential over-expression in Sensitive and Intermediate cell lines. The expression of these 3 genes is highly correlated, with correlation coefficients of +0.77 or higher. Due to their similarity, a single pair gene EPDR1 was selected from Tables 2-4 to measure contrasting overexpression in Resistant cell lines. Including such anti-correlated Pair Genes in the assay provides auto-normalization in that both Sensitivity and Resistance are associated with high expression of one arm of the pair. By this mechanism, the assay does not depend upon low overall mRNA assay levels to define any class, but rather describes each by a pattern of relative expression of the Main Genes to their anticorrelated Pairs (i.e. a sum of signed t-scores on the log2 scale, with signs corresponding to the Fold Change Estimate). In Steps 2-5, additional pairs of genes were chosen based upon mechanism of action from a new list of those with significant associations to IC25 after adjustment for the cumulative sum of signed t-scores for genes identified in previous Steps. This stepwise procedure requires each new pair of genes to add additional predictive power to the Sensitivity Index. After Step 5, no more gene pairs were needed for IC25 prediction. In Step 6, a single additional pair was added for its ability to predict cell viability at maximum inhibition after adjusting for the cumulative Index based upon the previous 7 pairs of genes. BCL6 was added as a singleton without a corresponding pair based upon a mechanism of action rationale: it is not currently incorporated in the final Sensitivity Index, which is given by the sum of signed t-scores for log 2-scale expression of Gene Pairs 1-8. It may be incorporated explicitly into the index based upon clinical experience. For classification into Sensitive or Intermediate versus Resistant groups, a preliminary cutoff was chosen for the Sensitivity Index so as to maximize the overall correct classification rate. Alternate classification rules based upon the selected probes may be optimized later for clinical application.
Results and Analysis
To gain an understanding of the mechanism of action of anti-CD40 antibody, and to identify one or more predictive markers for the responsiveness of NHL patients to anti-CD40 antibody therapy, we tested the activity of anti-CD40 Ab.1 across a panel of 31 NHL cell lines and assessed cell viability in response to a titration of anti-CD40 antibody. The IC25 values highlighted in Table 1 from this experiment reveal that anti-CD40 antibody sensitized 10 cell lines with a reduction in cell viability at a concentration of <0.4 μg/ml, hereon defined as ‘sensitive’ cell lines, and 13 cell lines that did not achieve a reduction in cell viability even up to concentrations of 1 μg/ml, hereon defined as ‘resistant’ cell lines. 8 cell lines had an IC25 between >0.4 and <0.8, and will hereon be defined as ‘intermediate’ cell lines.
Table 1 provides anti-CD40 Ab.1 IC25 sensitivity data across NHL cell lines in vitro. Specific lymphoma subtypes of each cell line, IC25 values and classifier data are given for each cell line. DLBCL (Diffuse Large B-cell Lymphoma), FL (Follicular Lymphoma, MCL (Mantle Cell Lymphoma), ALCL (Anaplastic Large Cell Lymphoma).
TABLE 1
Anti-CD40 Anti-CD40
Antibody Antibody
Sensitivity IC25
Cell line IC25 Classifier (μg/ml) Lymphoma Subtype
SU-DHL-16 Sensitive 0.009817124 DLBCL
SU-DHL-10 Sensitive 0.01 DLBCL
SU-DHL-8 Sensitive 0.011140955 DLBCL
SU-DHL-5 Sensitive 0.015309599 DLBCL
SU-DHL-4 Sensitive 0.03 DLBCL
MC116 Sensitive 0.03217012 UBCL
HT Sensitive 0.123333333 DLBCL
KARPAS- Sensitive 0.196666667 DLBCL
1106P
BJAB Sensitive 0.240995143 Burkitt's Lymphoma
WSU-NHL Sensitive 0.348838607 FL
REC-1 Intermediate 0.42 MCL
WSU-FSCCL Intermediate 0.49 FL
A3/Kawakami Intermediate 0.668463355 DLBCL
DB Intermediate 0.676933804 DLBCL
Ri-1 Intermediate 0.696666667 DLBCL
RL Intermediate 0.698508885 DLBCL
Sc-1 Intermediate 0.709276746 FL
Farage Intermediate 0.796666667 DLBCL
A4/Fukada Resistant 1 DLBCL
GRANTA-519 Resistant 1 MCL
JeKo-1 Resistant 1 MCL
Karpas-422 Resistant 1 DLBCL
NU-DHL-1 Resistant 1 DLBCL
OCI-Ly19 Resistant 1 DLBCL
Pfeiffer Resistant 1 DLBCL
RC-K8 Resistant 1 DLBCL
SCC-3 Resistant 1 DLBCL
SR-786 Resistant 1 ALCL
SU-DHL-1 Resistant 1 ALCL
TK Resistant 1 DLBCL
Toledo Resistant 1 DLBCL
To identify genes that are predictive of anti-CD40 Ab.1 activity in vitro, RNA was prepared from the cell lines at the log stage of cell division and subjected to gene expression profiling using the Affymetrix HGU133P2 microarray. Differentially expressed genes between Sensitive and Resistant cell lines were determined by a moderated t-test and significance was determined using an adjusted P-value cutoff of ≦0.05 (Table 2). In Table 2, gene list filtered to an adjusted p-value <0.05 (5% FDR) resulting in 110 unique genes. Probe ID, gene symbol and description are indicated. In addition, significant genes that correlated with the IC25 values across all NHL cell lines were determined by the Spearman's Rank Correlation and genes were filtered using a rho value of ≦−0.57 or ≧0.57 (Table 3). In Table 3, gene list filtered with a rho value of ≦−0.57 or ≧0.57 resulting in 130 unique genes. Probe ID, gene symbol and description are also indicated. A combined table of unique genes identified by each or both methodologies is displayed in Table 4. In Table 4, the Log(2) fold change is indicated where a positive fold change represents increased expression in the sensitive class and a negative fold change represents increased expression in the resistant class of NHL cell lines with respect to anti-CD40 Ab.1 sensitivity. Gene represents 195 unique genes. Probe IDs, gene symbol and description are also indicated.
TABLE 2
Gene Symbol Probe Description adj. P. Val
RGS13 210258_at regulator of G-protein signalling 13 2.57E−05
MGC2463 219812_at 0.00015799
VNN2 205922_at vanin 2 0.000247994
EPDR1 223253_at ependymin related protein 1 (zebrafish) 0.000434413
MEF2B 205124_at MADS box transcription enhancer factor 0.001352572
2, polypeptide B (myocyte enhancer factor
2B)
SLAMF6 1552497_a_at SLAM family member 6 0.00263509
LCK 204891_s_at lymphocyte-specific protein tyrosine 0.00263509
kinase
LPP 202822_at LIM domain containing preferred 0.005668066
translocation partner in lipoma
SLC30A1 212907_at solute carrier family 30 (zinc transporter), 0.00783662
member 1
LTB 207339_s_at lymphotoxin beta (TNF superfamily, 0.008947887
member 3)
FAM113B 228298_at family with sequence similarity 113, 0.008947887
member B
BRDG1 220059_at 0.011013653
PRPSAP2 203537_at phosphoribosyl pyrophosphate synthetase- 0.011342898
associated protein 2
244040_at 244040_at 0.011342898
SEMA4A 219259_at sema domain, immunoglobulin domain 0.012794771
(Ig), transmembrane domain (TM) and
short cytoplasmic domain, (semaphorin)
4A
CD86 210895_s_at CD86 molecule 0.013430782
CD22 217422_s_at CD22 molecule 0.01483858
LIMD1 222762_x_at LIM domains containing 1 0.01483858
236126_at 236126_at 0.01483858
RUNDC2B 1554411_s_at RUN domain containing 2B 0.01483858
LOXL2 202998_s_at lysyl oxidase-like 2 0.015908888
GOLPH2 217771_at golgi phosphoprotein 2 0.015908888
RASGRP3 205801_s_at RAS guanyl releasing protein 3 (calcium 0.015908888
and DAG-regulated)
C21orf7 221211_s_at chromosome 21 open reading frame 7 0.016054465
RAP1A 202362_at RAP1A, member of RAS oncogene family 0.016642805
ANKRD13A 224810_s_at ankyrin repeat domain 13A 0.016798331
ZNF32 209538_at zinc finger protein 32 0.017041183
DAAM1 216060_s_at dishevelled associated activator of 0.017041183
morphogenesis 1
CRTC3 218648_at CREB regulated transcription coactivator 3 0.017041183
C13orf31 228937_at chromosome 13 open reading frame 31 0.017041183
SMAP1L 225282_at stromal membrane-associated protein 1- 0.017041183
like
224811_at 224811_at 0.017041183
KCNN3 205903_s_at potassium intermediate/small conductance 0.017041183
calcium-activated channel, subfamily N,
member 3
S100Z 1554876_a_at S100 calcium binding protein, zeta 0.017041183
FZD1 204451_at frizzled homolog 1 (Drosophila) 0.017041183
FLVCR 222906_at 0.017041183
MYBL1 213906_at v-myb myeloblastosis viral oncogene 0.017041183
homolog (avian)-like 1
EHBP1 212653_s_at EH domain binding protein 1 0.017041183
SYNE2 242774_at spectrin repeat containing, nuclear 0.018508325
envelope 2
FLJ36492 1557366_at 0.018508325
MAP2K1 202670_at mitogen-activated protein kinase 1 0.018508325
NEIL1 219396_s_at nei endonuclease VIII-like 1 (E. coli) 0.018534278
228191_at 228191_at 0.018813942
LOC30203 225014_at 0.02072242
OPN3 219032_x_at opsin 3 (encephalopsin, panopsin) 0.021965295
227539_at 227539_at 0.022123902
GCHFR 204867_at GTP cyclohydrolase I feedback regulator 0.024418721
239287_at 239287_at 0.024681541
B3GALNT2 226233_at beta-1,3-N- 0.024681541
acetylgalactosaminyltransferase 2
ANUBL1 223624_at AN1, ubiquitin-like, homolog (Xenopus 0.024681541
laevis)
241879_at 241879_at 0.026428191
HDAC1 201209_at histone deacetylase 1 0.027641246
FHL1 201540_at four and a half LIM domains 1 0.027802063
PON2 201876_at paraoxonase 2 0.028969668
DNMT1 227684_at DNA (cytosine-5-)-methyltransferase 1 0.030015625
GABARAPL2 209046_s_at GABA(A) receptor-associated protein-like 2 0.031517586
HSP90B1 216449_x_at heat shock protein 90 kDa beta (Grp94), 0.031894346
member 1
RRAS2 212590_at related RAS viral (r-ras) oncogene 0.032663885
homolog 2
ARSG 230748_at arylsulfatase G 0.03380232
UGDH 203343_at UDP-glucose dehydrogenase 0.03380232
KCNMB4 222857_s_at potassium large conductance calcium- 0.03380232
activated channel, subfamily M, beta
member 4
SYTL1 227134_at synaptotagmin-like 1 0.034025836
CYFIP1 208923_at cytoplasmic FMR1 interacting protein 1 0.035718667
HIPK2 225368_at homeodomain interacting protein kinase 2 0.035718667
MAN2A2 202032_s_at mannosidase, alpha, class 2A, member 2 0.035718667
AAK1 225522_at AP2 associated kinase 1 0.035782217
TBPL1 208398_s_at TBP-like 1 0.036337106
1553979_at 1553979_at 0.037283374
CHML 226350_at choroideremia-like (Rab escort protein 2) 0.037979419
VARS 201796_s_at valyl-tRNA synthetase 0.037979419
PTK2 208820_at PTK2 protein tyrosine kinase 2 0.037979419
IGF1R 203627_at insulin-like growth factor 1 receptor 0.037979419
GRB2 215075_s_at growth factor receptor-bound protein 2 0.039960264
ATP8A1 213106_at ATPase, aminophospholipid transporter 0.039960264
(APLT), Class I, type 8A, member 1
FZD3 219683_at frizzled homolog 3 (Drosophila) 0.041405941
KIF1B 225878_at kinesin family member 1B 0.041405941
UBXD2 212008_at UBX domain containing 2 0.041405941
TMEM87A 212202_s_at transmembrane protein 87A 0.041888206
PARVB 37965_at parvin, beta 0.042377536
SLC26A2 205097_at solute carrier family 26 (sulfate 0.042377536
transporter), member 2
FCRLM1 235400_at Fc receptor-like and mucin-like 1 0.042377536
PDGFD 219304_s_at platelet derived growth factor D 0.043219716
PRDX4 201923_at peroxiredoxin 4 0.043219716
SERPINA9 1553499_s_at serpin peptidase inhibitor, clade A (alpha- 0.043248911
1 antiproteinase, antitrypsin), member 9
C6orf62 222309_at chromosome 6 open reading frame 62 0.043554388
226525_at 226525_at 0.043554388
TOB1 228834_at transducer of ERBB2, 1 0.043554388
228242_at 228242_at 0.043742426
PKHD1L1 230673_at polycystic kidney and hepatic disease 1 0.04395172
(autosomal recessive)-like 1
KLHL6 1560396_at kelch-like 6 (Drosophila) 0.04395172
ASB2 227915_at ankyrin repeat and SOCS box-containing 2 0.044799524
PLEKHF2 222699_s_at pleckstrin homology domain containing, 0.046489788
family F (with FYVE domain) member 2
KLHL23 213610_s_at kelch-like 23 (Drosophila) 0.046489788
CPNE2 225129_at copine II 0.046489788
LOC642236 215160_x_at 0.047687714
GALNT2 217787_s_at UDP-N-acetyl-alpha-D- 0.047687714
galactosamine:polypeptide N-
acetylgalactosaminyltransferase 2
(GalNAc-T2)
CD180 206206_at CD180 molecule 0.047687714
CPNE5 227189_at copine V 0.047687714
FH 203032_s_at fumarate hydratase 0.047687714
KIF14 206364_at kinesin family member 14 0.047687714
PEA15 200787_s_at phosphoprotein enriched in astrocytes 15 0.047687714
TOX 204529_s_at 0.047687714
MRPS31 212604_at mitochondrial ribosomal protein S31 0.047687714
SEC23A 204344_s_at Sec23 homolog A (S. cerevisiae) 0.047687714
DPYD 204646_at dihydropyrimidine dehydrogenase 0.047864579
227107_at 227107_at 0.047864579
RAB11FIP1 219681_s_at RAB11 family interacting protein 1 (class 0.047864579
I)
C1orf107 214193_s_at chromosome 1 open reading frame 107 0.047864579
ATXN10 208833_s_at ataxin 10 0.048252462
CPEB4 224831_at cytoplasmic polyadenylation element 0.048504075
binding protein 4
TABLE 3
Symbol Probe Description rho
SLC30A1 228181_at solute carrier family 30 (zinc 0.754838311
transporter), member 1
EPDR1 223253_at ependymin related protein 1 (zebrafish) 0.733893852
FZD1 204451_at frizzled homolog 1 (Drosophila) 0.732218295
MAN2A2 202032_s_at mannosidase, alpha, class 2A, member 2 0.721327176
PVRIG 219812_at −0.715881617
EHBP1 212653_s_at EH domain binding protein 1 0.706666055
DAAM1 226666_at G protein-coupled receptor 135 −0.705409387
SMAP1L 225282_at stromal membrane-associated protein 1- −0.704990498
like
PRPSAP2 203537_at phosphoribosyl pyrophosphate −0.702896052
synthetase-associated protein 2
HSP90B1 216449_x_at heat shock protein 90 kDa beta (Grp94), 0.691586044
member 1
ZNF322A 219376_at zinc finger protein 322A 0.690748265
TMEM87A 212202_s_at transmembrane protein 87A 0.68823493
RABGAP1L 213982_s_at RAB GTPase activating protein 1-like −0.681951593
EAF2 219551_at ELL associated factor 2 −0.681532703
KCNMB4 234034_at potassium large conductance calcium- −0.673992698
activated channel, subfamily M, beta
member 4
LCK 204891_s_at lymphocyte-specific protein tyrosine −0.668547139
kinase
RGS13 1568752_s_at regulator of G-protein signalling 13 −0.666452693
TOB1 228834_at transducer of ERBB2, 1 −0.663520468
PLEKHF2 218640_s_at pleckstrin homology domain containing, −0.66268269
family F (with FYVE domain) member 2
TBPL1 208398_s_at TBP-like 1 −0.658912687
KLHL23 230434_at kelch-like 23 (Drosophila) 0.658493798
SEMA4C 46665_at sema domain, immunoglobulin domain 0.658074909
(Ig), transmembrane domain (TM) and
short cytoplasmic domain, (semaphorin)
4C
CRTC3 218648_at CREB regulated transcription 0.657237131
coactivator 3
237075_at 237075_at −0.657237131
GCS1 210627_s_at 0.650534904
CPNE2 225129_at copine II 0.642576009
PIGL 205873_at phosphatidylinositol glycan anchor −0.64215712
biosynthesis, class L
MTHFR 239035_at 5,10-methylenetetrahydrofolate −0.64215712
reductase (NADPH)
ENTPD6 201704_at ectonucleoside triphosphate 0.641319342
diphosphohydrolase 6 (putative
function)
CD22 204581_at CD22 molecule −0.640062674
TPD52 201691_s_at tumor protein D52 −0.637549339
GPSM1 226043_at G-protein signalling modulator 1 0.633360447
(AGS3-like, C. elegans)
239467_at 239467_at −0.632941558
ROCK1 213044_at Rho-associated, coiled-coil containing −0.632522669
protein kinase 1
CENTB2 212476_at centaurin, beta 2 −0.630847112
WIPF1 231182_at Wiskott-Aldrich syndrome protein −0.629590445
interacting protein
RAB11FIP1 219681_s_at RAB11 family interacting protein 1 −0.628333777
(class I)
LPP 202822_at LIM domain containing preferred −0.627077109
translocation partner in lipoma
FLJ22814 220674_at −0.62665822
TRAP1 228929_at TNF receptor-associated protein 1 −0.62665822
MRPS31 212603_at mitochondrial ribosomal protein S31 −0.625401553
ANKRD13A 224810_s_at ankyrin repeat domain 13A −0.625401553
GALNT2 217788_s_at UDP-N-acetyl-alpha-D- 0.624982664
galactosamine:polypeptide N-
acetylgalactosaminyltransferase 2
(GalNAc-T2)
ACVR2B 236126_at 0.623160484
CD180 206206_at CD180 molecule −0.62163155
IXL 225708_at intersex-like (Drosophila) 0.62163155
FAM113B 228298_at family with sequence similarity 113, −0.621212661
member B
MEF2B 205124_at MADS box transcription enhancer factor −0.620793772
2, polypeptide B (myocyte enhancer
factor 2B)
224811_at 224811_at −0.620374882
ATP6V1A 201972_at ATPase, H+ transporting, lysosomal −0.619955993
70 kDa, V1 subunit A
SLC15A2 205316_at solute carrier family 15 (H+/peptide −0.618280437
transporter), member 2
RTN4IP1 224509_s_at reticulon 4 interacting protein 1 −0.618280437
TTC9 213174_at tetratricopeptide repeat domain 9 −0.615767101
PTPRC 212587_s_at protein tyrosine phosphatase, receptor −0.615348212
type, C
FLJ43663 228702_at −0.615348212
MARCH6 201736_s_at membrane-associated ring finger 0.615348212
(C3HC4) 6
C13orf31 228937_at chromosome 13 open reading frame 31 −0.614929323
CNOT6L 226153_s_at CCR4-NOT transcription complex, −0.614091545
subunit 6-like
PIGW 1558292_s_at phosphatidylinositol glycan anchor 0.61115932
biosynthesis, class W
ARTS-1 210385_s_at 0.610740431
RYK 216976_s_at RYK receptor-like tyrosine kinase 0.609483764
VNN2 205922_at vanin 2 −0.609483764
FNTB 204764_at farnesyltransferase, CAAX box, beta 0.608645985
BICD1 242052_at bicaudal D homolog 1 (Drosophila) −0.607808207
SEPT8 209000_s_at septin 8 0.606970429
WDR6 233573_s_at WD repeat domain 6 0.606551539
HDAC1 201209_at histone deacetylase 1 −0.604038204
ATP2B4 212135_s_at ATPase, Ca++ transporting, plasma 0.604038204
membrane 4
BRDG1 220059_at −0.602781537
SERPINA9 1553499_s_at serpin peptidase inhibitor, clade A −0.602362648
(alpha-1 antiproteinase, antitrypsin),
member 9
CRSP6 221517_s_at cofactor required for Sp1 transcriptional 0.602362648
activation, subunit 6, 77 kDa
TMEM17 1557137_at transmembrane protein 17 0.602362648
BPNT1 232103_at 3′(2′), 5′-bisphosphate nucleotidase 1 −0.601943758
242826_at 242826_at −0.601524869
NCOA3 207700_s_at nuclear receptor coactivator 3 −0.598592645
LRMP 35974_at lymphoid-restricted membrane protein −0.598592645
PTK2 208820_at PTK2 protein tyrosine kinase 2 −0.598173756
C21orf7 221211_s_at chromosome 21 open reading frame 7 −0.598173756
FCRL3 231093_at Fc receptor-like 3 −0.598173756
FDFT1 208647_at farnesyl-diphosphate farnesyltransferase 1 −0.597335977
DHX38 209178_at DEAH (Asp-Glu-Ala-His) box 0.596917088
polypeptide 38
C1orf57 223272_s_at chromosome 1 open reading frame 57 0.596917088
ARSG 230748_at arylsulfatase G −0.595660421
MS4A7 223343_at membrane-spanning 4-domains, −0.595241531
subfamily A, member 7
CYP39A1 244407_at cytochrome P450, family 39, subfamily −0.594403753
A, polypeptide 1
DCK 203302_at deoxycytidine kinase −0.593565975
CTNNA1 1558214_s_at catenin (cadherin-associated protein), 0.593565975
alpha 1, 102 kDa
SLC27A2 205769_at solute carrier family 27 (fatty acid 0.592728196
transporter), member 2
SLC35B2 224716_at solute carrier family 35, member B2 0.592309307
243185_at 243185_at −0.592309307
FAM89B 32209_at family with sequence similarity 89, 0.591890418
member B
GSG2 223759_s_at germ cell associated 2 (haspin) −0.591471529
USP6NL 204761_at USP6 N-terminal like −0.59105264
ATPIF1 218671_s_at ATPase inhibitory factor 1 −0.590214861
SLAMF6 1552497_a_at SLAM family member 6 −0.590214861
TARSL2 227611_at threonyl-tRNA synthetase-like 2 0.590214861
XKR6 236047_at XK, Kell blood group complex subunit- −0.589377083
related family, member 6
228242_at 228242_at 0.588958194
EYA3 1552314_a_at eyes absent homolog 3 (Drosophila) −0.586863748
RUNDC2B 1554413_s_at RUN domain containing 2B −0.584350413
BXDC5 218462_at brix domain containing 5 −0.583512634
SLC26A2 205097_at solute carrier family 26 (sulfate 0.583512634
transporter), member 2
PNMA1 218224_at paraneoplastic antigen MA1 0.583512634
LOC401504 226635_at −0.583093745
GPR82 1553316_at G protein-coupled receptor 82 −0.582674856
ZBTB9 226163_at zinc finger and BTB domain containing 9 0.582255967
BFSP2 207399_at beaded filament structural protein 2, −0.580999299
phakinin
SLC6A16 219820_at solute carrier family 6, member 16 −0.580999299
SBNO2 204166_at KIAA0963 0.580161521
CTSC 201487_at cathepsin C 0.579323742
EID1 208669_s_at CREBBP/EP300 inhibitor 1 0.579323742
RRAS2 212589_at related RAS viral (r-ras) oncogene −0.578904853
homolog 2
NLK 238624_at nemo-like kinase −0.578904853
FLJ36492 1557366_at −0.578904853
RALGDS 209051_s_at ral guanine nucleotide dissociation 0.578485964
stimulator
CIRBP 225191_at cold inducible RNA binding protein 0.578067075
P4HB 1564494_s_at procollagen-proline, 2-oxoglutarate 4- 0.578067075
dioxygenase (proline 4-hydroxylase),
beta polypeptide
ATG3 221492_s_at ATG3 autophagy related 3 homolog (S. cerevisiae) −0.578067075
227539_at 227539_at −0.577648186
FLJ10815 56821_at 0.577648186
C19orf54 222052_at chromosome 19 open reading frame 54 −0.577229296
PORCN 219483_s_at porcupine homolog (Drosophila) 0.576810407
PDE6D 204091_at phosphodiesterase 6D, cGMP-specific, −0.576391518
rod, delta
LOC389203 225014_at −0.576391518
235018_at 235018_at −0.575134851
CDK10 210622_x_at cyclin-dependent kinase (CDC2-like) 10 0.575134851
KYNU 210662_at kynureninase (L-kynurenine hydrolase) −0.573878183
PIGG 218652_s_at phosphatidylinositol glycan anchor 0.573878183
biosynthesis, class G
TMEM64 225972_at transmembrane protein 64 −0.573878183
NEDD9 240019_at neural precursor cell expressed, −0.573878183
developmentally down-regulated 9
TABLE 4
Symbol Probes Description logFC Adj. P. value rho
EPDR1 223253_at ependymin related protein 1 −6.71079565 4.3441E−04 0.734
(zebrafish)
HIPK2 225368_at NA −5.568390135 3.5719E−02 NA
CYFIP1 208923_at NA −5.507430049 3.5719E−02 NA
GOLPH2 217771_at NA −5.149533123 1.5909E−02 NA
PON2 201876_at NA −5.02937768 2.8970E−02 NA
OPN3 219032_x_at NA −4.868576042 2.1965E−02 NA
FHL1 201540_at NA −4.849936383 2.7802E−02 NA
DPYD 204646_at NA −4.601899147 4.7865E−02 NA
CRTC3 218648_at CREB regulated −4.447380308 1.7041E−02 0.657
transcription coactivator 3
LIMD1 222762_x_at NA −4.385468009 1.4839E−02 NA
IGF1R 203627_at NA −3.780119703 3.7979E−02 NA
PARVB 37965_at NA −3.705700946 4.2378E−02 NA
236126_at 236126_at NA −3.694482091 1.4839E−02 NA
CHML 226350_at NA −3.643899135 3.7979E−02 NA
FZD1 204451_at frizzled homolog 1 −3.531407505 1.7041E−02 0.732
(Drosophila)
AAK1 225522_at NA −3.502784982 3.5782E−02 NA
CPNE2 225129_at copine II −3.432724459 4.6490E−02 0.643
KLHL23 213610_s_at, 230434_at kelch-like 23 (Drosophila) −3.407601857 4.6490E−02 0.658
ZNF32 209538_at NA −3.37444837 1.7041E−02 NA
GALNT2 217787_s_at, 217788_s_at UDP-N-acetyl-alpha-D- −3.068993195 4.7688E−02 0.625
galactosamine:polypeptide
N-
acetylgalactosaminyltransferase
2 (GalNAc-T2)
SLC30A1 212907_at, 228181_at solute carrier family 30 (zinc −2.897034114 7.8366E−03 0.755
transporter), member 1
KIF1B 225878_at NA −2.893360476 4.1406E−02 NA
FZD3 219683_at NA −2.888266087 4.1406E−02 NA
SLC26A2 205097_at solute carrier family 26 −2.592191782 4.2378E−02 0.584
(sulfate transporter), member 2
VARS 201796_s_at NA −2.146698292 3.7979E−02 NA
MAN2A2 202032_s_at mannosidase, alpha, class −2.05163539 3.5719E−02 0.721
2A, member 2
C6orf62 222309_at NA −1.970715812 4.3554E−02 NA
UGDH 203343_at NA −1.915040205 3.3802E−02 NA
HSP90B1 216449_x_at heat shock protein 90 kDa −1.779135947 3.1894E−02 0.692
beta (Grp94), member 1
B3GALNT2 226233_at NA −1.591532059 2.4682E−02 NA
FLVCR 222906_at NA −1.528203803 1.7041E−02 NA
227107_at 227107_at NA −1.436834856 4.7865E−02 NA
SEC23A 204344_s_at NA −1.377564142 4.7688E−02 NA
228242_at 228242_at NA −1.314847732 4.3742E−02 0.589
TMEM87A 212202_s_at transmembrane protein 87A −1.267840163 4.1888E−02 0.688
228191_at 228191_at NA −1.196685963 1.8814E−02 NA
KIF14 206364_at NA −1.150921894 4.7688E−02 NA
EHBP1 212653_s_at EH domain binding protein 1 −1.110923792 1.7041E−02 0.707
C1orf107 214193_s_at NA −1.102968299 4.7865E−02 NA
UBXD2 212008_at NA −1.062833934 4.1406E−02 NA
FH 203032_s_at NA −1.047497846 4.7688E−02 NA
PRDX4 201923_at NA −0.976330782 4.3220E−02 NA
1553979_at 1553979_at NA −0.95937263 3.7283E−02 NA
ATXN10 208833_s_at NA 0.717153159 4.8252E−02 NA
GABARAPL2 209046_s_at NA 0.928831609 3.1518E−02 NA
MAP2K1 202670_at NA 1.062284638 1.8508E−02 NA
LOC642236 215160_x_at NA 1.091751999 4.7688E−02 NA
MRPS31 212604_at, 212603_at mitochondrial ribosomal 1.140136013 4.7688E−02 −0.625
protein S31
HDAC1 201209_at histone deacetylase 1 1.189759283 2.7641E−02 −0.604
RAP1A 202362_at NA 1.235628621 1.6643E−02 NA
226525_at 226525_at NA 1.46297442 4.3554E−02 NA
TBPL1 208398_s_at TBP-like 1 1.50757518 3.6337E−02 −0.659
TOB1 228834_at transducer of ERBB2, 1 1.580519874 4.3554E−02 −0.664
SMAP1L 225282_at stromal membrane- 1.582665273 1.7041E−02 −0.705
associated protein 1-like
PEA15 200787_s_at NA 1.636511829 4.7688E−02 NA
LOC389203 225014_at NA 1.653219861 2.0722E−02 −0.576
227539_at 227539_at NA 1.706768556 2.2124E−02 −0.578
GRB2 215075_s_at NA 1.719009368 3.9960E−02 NA
PRPSAP2 203537_at phosphoribosyl 1.937200364 1.1343E−02 −0.703
pyrophosphate synthetase-
associated protein 2
ANKRD13A 224810_s_at ankyrin repeat domain 13A 2.096260555 1.6798E−02 −0.625
DAAM1 216060_s_at, 226666_at G protein-coupled receptor 2.205266761 1.7041E−02 −0.705
135
SYNE2 242774_at NA 2.326279517 1.8508E−02 NA
ATP8A1 213106_at NA 2.351268406 3.9960E−02 NA
PLEKHF2 222699_s_at, 218640_s_at pleckstrin homology domain 3.004500438 4.6490E−02 −0.663
containing, family F (with
FYVE domain) member 2
S100Z 1554876_a_at NA 3.144995156 1.7041E−02 NA
FLJ36492 1557366_at NA 3.222537979 1.8508E−02 −0.579
SLAMF6 1552497_a_at SLAM family member 6 3.363017096 2.6351E−03 −0.590
CPEB4 224831_at NA 3.444268629 4.8504E−02 NA
NEIL1 219396_s_at NA 3.470614786 1.8534E−02 NA
KLHL6 1560396_at NA 3.592234269 4.3952E−02 NA
ANUBL1 223624_at NA 3.597608491 2.4682E−02 NA
SYTL1 227134_at NA 3.601625514 3.4026E−02 NA
LPP 202822_at LIM domain containing 3.65635503 5.6681E−03 −0.627
preferred translocation
partner in lipoma
ARSG 230748_at arylsulfatase G 3.772680821 3.3802E−02 −0.596
DNMT1 227684_at NA 3.787896364 3.0016E−02 NA
RAB11FIP1 219681_s_at RAB11 family interacting 3.877841023 4.7865E−02 −0.628
protein 1 (class I)
224811_at 224811_at NA 3.884011816 1.7041E−02 −0.620
241879_at 241879_at NA 3.897073844 2.6428E−02 NA
MYBL1 213906_at NA 3.964686033 1.7041E−02 NA
KCNN3 244040_at potassium large conductance 4.14713855 1.1343E−02 NA
calcium-activated channel,
subfamily M, beta member 3
RUNDC2B 1554413_s_at RUN domain containing 2B 4.249552511 1.4839E−02 −0.584
GCHFR 204867_at NA 4.314659424 2.4419E−02 NA
C13orf31 228937_at chromosome 13 open 4.342634637 1.7041E−02 −0.615
reading frame 31
KCNN3 205903_s_at NA 4.348558398 1.7041E−02 NA
SERPINA9 1553499_s_at serpin peptidase inhibitor, 4.362716185 4.3249E−02 −0.602
clade A (alpha-1
antiproteinase, antitrypsin),
member 9
ASB2 227915_at NA 4.393168852 4.4800E−02 NA
CD180 206206_at CD180 molecule 4.400474176 4.7688E−02 −0.622
SEMA4A 219259_at NA 4.461977712 1.2795E−02 NA
PKHD1L1 230673_at NA 4.462674523 4.3952E−02 NA
FAM113B 228298_at family with sequence 4.725746806 8.9479E−03 −0.621
similarity 113, member B
MGC2463 219812_at NA 4.747120819 1.5799E−04 NA
PTK2 208820_at PTK2 protein tyrosine 4.830737904 3.7979E−02 −0.598
kinase 2
LTB 207339_s_at NA 4.861032521 8.9479E−03 NA
LOXL2 202998_s_at NA 4.936851624 1.5909E−02 NA
KCNMB4 222857_s_at, 234034_at potassium large conductance 5.103201059 3.3802E−02 −0.674
calcium-activated channel,
subfamily M, beta member 4
PDGFD 219304_s_at NA 5.13661915 4.3220E−02 NA
CD22 217422_s_at, 204581_at CD22 molecule 5.283886004 1.4839E−02 −0.640
CPNE5 227189_at NA 5.346723772 4.7688E−02 NA
C21orf7 221211_s_at chromosome 21 open 5.407994478 1.6054E−02 −0.598
reading frame 7
CD86 210895_s_at NA 5.574519784 1.3431E−02 NA
VNN2 205922_at vanin 2 5.634272247 2.4799E−04 −0.609
TOX 204529_s_at NA 5.647082288 4.7688E−02 NA
RASGRP3 205801_s_at NA 5.676809838 1.5909E−02 NA
RRAS2 212590_at, 212589_at related RAS viral (r-ras) 5.694136051 3.2664E−02 −0.579
oncogene homolog 2
239287_at 239287_at NA 5.91276116 2.4682E−02 NA
MEF2B 205124_at MADS box transcription 6.009095593 1.3526E−03 −0.621
enhancer factor 2,
polypeptide B (myocyte
enhancer factor 2B)
BRDG1 220059_at NA 6.358345958 1.1014E−02 −0.603
FCRLM1 235400_at NA 6.390558096 4.2378E−02 NA
LCK 204891_s_at lymphocyte-specific protein 7.315280882 2.6351E−03 −0.669
tyrosine kinase
RGS13 210258_at, 1568752_s_at regulator of G-protein 10.29738517 2.5700E−05 −0.666
signalling 13
PVRIG 219812_at NA NA NA −0.716
RABGAP1L 213982_s_at RAB GTPase activating NA NA −0.682
protein 1-like
EAF2 219551_at ELL associated factor 2 NA NA −0.682
237075_at 237075_at NA NA NA −0.657
MTHFR 239035_at 5,10- NA NA −0.642
methylenetetrahydrofolate
reductase (NADPH)
PIGL 205873_at phosphatidylinositol glycan NA NA −0.642
anchor biosynthesis, class L
TPD52 201691_s_at tumor protein D52 NA NA −0.638
239467_at 239467_at NA NA NA −0.633
ROCK1 213044_at Rho-associated, coiled-coil NA NA −0.633
containing protein kinase 1
CENTB2 212476_at centaurin, beta 2 NA NA −0.631
WIPF1 231182_at Wiskott-Aldrich syndrome NA NA −0.630
protein interacting protein
FLJ22814 220674_at NA NA NA −0.627
TRAP1 228929_at TNF receptor-associated NA NA −0.627
protein 1
ATP6V1A 201972_at ATPase, H+ transporting, NA NA −0.620
lysosomal 70 kDa, V1
subunit A
RTN4IP1 224509_s_at reticulon 4 interacting NA NA −0.618
protein 1
SLC15A2 205316_at solute carrier family 15 NA NA −0.618
(H+/peptide transporter),
member 2
TTC9 213174_at tetratricopeptide repeat NA NA −0.616
domain 9
FLJ43663 228702_at NA NA NA −0.615
PTPRC 212587_s_at protein tyrosine phosphatase, NA NA −0.615
receptor type, C
CNOT6L 226153_s_at CCR4-NOT transcription NA NA −0.614
complex, subunit 6-like
BICD1 242052_at bicaudal D homolog 1 NA NA −0.608
(Drosophila)
BPNT1 232103_at 3′(2′),5′-bisphosphate NA NA −0.602
nucleotidase 1
KAR 242826_at 3-ketoacyl-CoA reductase NA NA −0.602
LRMP 35974_at lymphoid-restricted NA NA −0.599
membrane protein
NCOA3 207700_s_at nuclear receptor coactivator 3 NA NA −0.599
FCRL3 231093_at Fc receptor-like 3 NA NA −0.598
FDFT1 208647_at farnesyl-diphosphate NA NA −0.597
farnesyltransferase 1
MS4A7 223343_at membrane-spanning 4- NA NA −0.595
domains, subfamily A,
member 7
CYP39A1 244407_at cytochrome P450, family 39, NA NA −0.594
subfamily A, polypeptide 1
DCK 203302_at deoxycytidine kinase NA NA −0.594
243185_at 243185_at NA NA NA −0.592
GSG2 223759_s_at germ cell associated 2 NA NA −0.591
(haspin)
USP6NL 204761_at USP6 N-terminal like NA NA −0.591
ATPIF1 218671_s_at ATPase inhibitory factor 1 NA NA −0.590
XKR6 236047_at XK, Kell blood group NA NA −0.589
complex subunit-related
family, member 6
EYA3 1552314_a_at eyes absent homolog 3 NA NA −0.587
(Drosophila)
BXDC5 218462_at brix domain containing 5 NA NA −0.584
LOC401504 226635_at NA NA NA −0.583
GPR82 1553316_at G protein-coupled receptor NA NA −0.583
82
BFSP2 207399_at beaded filament structural NA NA −0.581
protein 2, phakinin
SLC6A16 219820_at solute carrier family 6, NA NA −0.581
member 16
NLK 238624_at nemo-like kinase NA NA −0.579
ATG3 221492_s_at ATG3 autophagy related 3 NA NA −0.578
homolog (S. cerevisiae)
C19orf54 222052_at chromosome 19 open NA NA −0.577
reading frame 54
PDE6D 204091_at phosphodiesterase 6D, NA NA −0.576
cGMP-specific, rod, delta
235018_at 235018_at NA NA NA −0.575
KYNU 210662_at kynureninase (L-kynurenine NA NA −0.574
hydrolase)
NEDD9 240019_at neural precursor cell NA NA −0.574
expressed, developmentally
down-regulated 9
TMEM64 225972_at transmembrane protein 64 NA NA −0.574
PIGG 218652_s_at phosphatidylinositol glycan NA NA 0.574
anchor biosynthesis, class G
CDK10 210622_x_at cyclin-dependent kinase NA NA 0.575
(CDC2-like) 10
PORCN 219483_s_at porcupine homolog NA NA 0.577
(Drosophila)
FLJ10815 56821_at NA NA NA 0.578
CIRBP 225191_at cold inducible RNA binding NA NA 0.578
protein
P4HB 1564494_s_at procollagen-proline, 2- NA NA 0.578
oxoglutarate 4-dioxygenase
(proline 4-hydroxylase), beta
polypeptide
RALGDS 209051_s_at ral guanine nucleotide NA NA 0.578
dissociation stimulator
CTSC 201487_at cathepsin C NA NA 0.579
EID1 208669_s_at CREBBP/EP300 inhibitor 1 NA NA 0.579
SBNO2 204166_at KIAA0963 NA NA 0.580
ZBTB9 226163_at zinc finger and BTB domain NA NA 0.582
containing 9
PNMA1 218224_at paraneoplastic antigen MA1 NA NA 0.584
TARSL2 227611_at threonyl-tRNA synthetase- NA NA 0.590
like 2
FAM89B 32209_at family with sequence NA NA 0.592
similarity 89, member B
SLC35B2 224716_at solute carrier family 35, NA NA 0.592
member B2
SLC27A2 205769_at solute carrier family 27 NA NA 0.593
(fatty acid transporter),
member 2
CTNNA1 1558214_s_at catenin (cadherin-associated NA NA 0.594
protein), alpha 1, 102 kDa
C1orf57 223272_s_at chromosome 1 open reading NA NA 0.597
frame 57
DHX38 209178_at DEAH (Asp-Glu-Ala-His) NA NA 0.597
box polypeptide 38
CRSP6 221517_s_at cofactor required for Sp1 NA NA 0.602
transcriptional activation,
subunit 6, 77 kDa
TMEM17 1557137_at transmembrane protein 17 NA NA 0.602
ATP2B4 212135_s_at ATPase, Ca++ transporting, NA NA 0.604
plasma membrane 4
WDR6 233573_s_at WD repeat domain 6 NA NA 0.607
SEPT8 209000_s_at septin 8 NA NA 0.607
FNTB 204764_at farnesyltransferase, CAAX NA NA 0.609
box, beta
RYK 216976_s_at RYK receptor-like tyrosine NA NA 0.609
kinase
ARTS-1 210385_s_at NA NA NA 0.611
PIGW 1558292_s_at phosphatidylinositol glycan NA NA 0.611
anchor biosynthesis, class W
MARCH6 201736_s_at membrane-associated ring NA NA 0.615
finger (C3HC4) 6
IXL 225708_at intersex-like (Drosophila) NA NA 0.622
ACVR2B 236126_at NA NA NA 0.623
GPSM1 226043_at G-protein signalling NA NA 0.633
modulator 1 (AGS3-like, C. elegans)
ENTPD6 201704_at ectonucleoside triphosphate NA NA 0.641
diphosphohydrolase 6
(putative function)
GCS1 210627_s_at NA NA NA 0.651
SEMA4C 46665_at sema domain, NA NA 0.658
immunoglobulin domain
(Ig), transmembrane domain
(TM) and short cytoplasmic
domain, (semaphorin) 4C
ZNF322A 219376_at zinc finger protein 322A NA NA 0.691
The genes that are highly expressed in Table 4 may be co-regulated genes that may not be related to the biology of anti-CD40 activity. Therefore to comprehend the biological function of the genes that are differentially expressed between sensitive and resistant cells, we carried out Gene Set Enrichment Analysis (GSEA). In this analysis, we address the question by calculating the mean t-statistic for genes in the set, and then comparing that mean t-statistic to the mean statistics calculated for random sets of genes of the same size. A low p-value may indicate that there is some correlation between the set of genes and the sample classification used to generate the statistics. Gene Set Analysis can thus be interpreted as a summary of the properties of the genes that are highly differentially expressed. Table 5 provides gene set enrichment analysis of anti-CD40 Ab.1 Sensitive vs. Resistant NHL cell lines. Enriched gene sets, number of genes per gene set, normalized enrichment score (NES), and nominal p-value (NOM p-val) are displayed. The higher the NES and the lower the NOM p-val, the more likely the findings are significant.
TABLE 5
Number of
Gene Set Name Genes NES NOM p-val
BCRPATHWAY 35 1.5387669 0.018181818
BASSO_GERMINAL_CENTER- 70 1.5124674 0.016949153
CD40_DN
Of the GSEA identified gene sets that were biologically relevant, gene sets involved in B-cell Receptor Signaling (BCR) and genes that are of germinal center origin (Table 5) were enriched. Of primary interest is the observation of genes involved in CD40 signaling as determined by the BASSO_GERMINAL_CENTER_CD40DN gene set (FIG. 1). Basso et al., Blood 104:4088-96, 2004. This gene set refers to genes that have been reported to be repressed by CD40L in a Ramos cell line. The rank and adjusted p-value from the differentially expressed gene list is displayed in Table 6 with respect to this gene set. In Table 6, differentially expressed genes between sensitive and resistant cell lines are enriched for genes that are known to be CD40L downregulated. Ranked genes are derived from the moderated t-test (Table 2). 70 genes in total were part of this gene set with the top 11 being displayed in this table. Genes shown in table 6 were overexpressed in anti-CD40 Ab.1 sensitive cell lines. The partial overlap of genes with the BCR and CD40L genes is expected since the two signal transduction pathways converge at the axis of NF-κB transcription and both pathways can synergize to activate B-cells. We next ascertained if any of the CD40L-induced genes are capable of discriminating between sensitive and resistant NHL cell lines to anti-CD40 Ab.1. Of the CD40L genes within the differentially expressed gene list on Tables 2 and 3, VNN2 gave the most accurate discrimination for sensitive and resistant cell lines (FIG. 2).
TABLE 6
Gene
Rank Symbol ProbeID Description t-statistic pvalue adj. P. Val.
3 VNN2 205922_at vanin 2 7.2679 0 0.000248
5 MEF2C 205124_at MADS box transcription 6.7125 0 0.001353
enhancer factor 2,
polypeptide B (myocyte
enhancer factor 2B)
10 LTB 207339_s_at lymphotoxin beta (TNF 4.8723 1.00E−04 0.008948
superfamily, member 3)
14 KCNN3 244040_at potassium 5.4914 0 0.011343
intermediate/small
conductance calcium-
activated channel,
subfamily N, member 3
252 NCF1 204961_s_at NCF1 4.0453 6.00E−04 0.094030
278 BCL6 203140_at B-cell CLL/lymphoma 6 4.3355 3.00E−04 0.098016
(zinc finger protein 51)
349 IGJ 212592_at immunoglobulin J 3.6952 0.0013 0.109865
polypeptide, linker protein
for immunoglobulin alpha
and mu polypeptides
475 ELTI1902 207761_s_at methyltransferase like 7A 3.3433 0.0031 0.130104
498 PNOC 205901_at prepronociceptin 3.7812 0.0011 0.134773
548 CSF2RB 205159_at colony stimulating factor 2 3.3371 0.0031 0.146260
receptor, beta, low-affinity
(granulocyte-macrophage)
707 POU2AF1 205267_at POU domain, class 2, 3.3788 0.0028 0.171312
associating factor 1
Further inspection of the differentially expressed genes list also revealed genes such as CD22, RGS13, and MEF2B (Table 2 and FIGS. 3, 4, 6), that were indicative of germinal center B (GCB) cells were overexpressed in anti-CD40 Ab.1 sensitive cell lines. CD40 signature genes correlated with anti-CD40.Ab.1 sensitivity as shown in FIG. 5. Notably, RGS13 was one of the highest-ranking genes by moderated t-test (Table 2) and Spearman's rank correlation (Table 3) across the cell lines and as a single gene can discriminate between sensitive and resistant as well intermediate and resistant classes with high accuracy: 96% accuracy for sensitive vs. resistant. 81% for intermediate vs. resistant, and 87% for sensitive/intermediate vs. resistant.
To gain optimal classification accuracy it will likely require a gene signature, or metagene, classifier. Therefore, to identify genes that may contribute to the most accurate classifier we generated an algorithm to identify pairs of genes that when combined would give the best possible classification across the cell lines with respect to anti-CD40 Ab.1 sensitivity. We therefore carried out a Stepwise Linear Modeling to achieve this aim and the final gene selection is shown in Table 7. In Table 7, each target gene is shown with its corresponding inversely correlated (anti-correlated) Pair Gene, in order of the step at which the Target Gene was chosen for inclusion in the Index, as described earlier. This selection of gene pairs revealed a robust classification of Sensitive, Intermediate and Resistant classes to anti-CD40 Ab.1 (FIG. 4) when a Sensitivity Index was calculated, which is essentially the sum of signed t-scores for log 2-scale expression of Gene Pairs 1-8.
TABLE 7
Anti-CD40 Ab.1 Sensitivity Index Identified Using Stepwise Linear Modeling.
Gene Main Fold Correlation
Pair Step Gene Main Gene Change Pair Gene Pair Gene with Main
# # Symbol Probe Estimate Symbol Probe Gene
1 1 VNN2 205922_at +2.63 EPDR1 223253_at −0.72
2 1 RGS13 210258_at +5.18 EPDR1 223253_at −0.88
3 1 CD22 204581_at +2.70 EPDR1 223253_at −0.68
4 2 LRRC8A 233487_s_at −0.50 PRPSAP2 203537_at −0.61
5 3 CD40 205153_s_at +1.47 IGF1R 203627_at −0.76
6 4 IFITM1 214022_s_at −2.01 BTG2 201236_s_at −0.56
7 5 SMN1 203852_s_at +0.36 LMO2 204249_s_at −0.49
8 6 PRKCA 213093_at −1.34 YIPF3 216338_s_at −0.72
9 7 BCL6 203140_at NA NA NA NA
Overall, CD40L plays a critical role in activating B-cells and results in the expansion and proliferation of B-cells as well as Ig class switching and the CD40L signaling pathway is also active within pre- and post-GCB-cells including naïve and memory B-cells. Therefore, it is striking to note that NHL cells that are displaying sensitivity to anti-CD40 Ab.1 are similar to GCB-cells in origin by gene expression profiling and have CD40L downregulated genes highly expressed, in contrast to resistant cells, indicative of a relationship between GCB and CD40 pathway activation status determining sensitivity to anti-CD40 Ab. 1.
To further confirm predictive classifier, xenograft models are used to explore in therapy (such as combination therapy). Real time quantitative RT-PCR (qRT-PCR) is used for measuring gene expression levels. After confirming the predictive classifier, immunohistochemistry (IHC) assays are developed for a small group of markers selected (e.g., VNN2 and RGS13). Selected marker genes are further tested in clinical trial samples.
qRT-PCR and IHC are performed to measure expression levels of selected marker genes in clinical trial samples. Expression levels in samples from patients having relapsed diffuse large B-cell lymphoma that are responsive to the anti-CD40 treatment are compared the expression levels in samples from patients that are not responsive to the treatment.
Example 2 Identification of Markers Associated with Responsiveness to Treatment with Anti-CD40 Ab.1 in Clinical Trials
Clinical Trial 001 (Phase II)
A multicenter, phase II, open-label study to determine the overall response rate and toxicity profile of anti-CD40 Ab.1 in patients with relapsed DLBCL. Tumor samples were assessed by a central lab for pathology confirmation and CD40 expression. Eligible patients had de novo or a transformed DLBCL at diagnosis and were excluded if there was a prior history of indolent lymphoma. Required prior therapy consisted of combination chemotherapy with rituximab and, if eligible, autologous stem cell transplantation. Patients received 6 IV infusions of anti-CD40 Ab.1 over 5 weeks (Cycle 1) with intra-patient dose loading (1 mg/kg on Day 1; 2 mg/kg on Day 4; 4 mg/kg on Day 8) and 8 mg/kg/wk thereafter. Responding patients and those with SD (stable disease) were eligible to continue therapy until disease progression or up to a maximum of 12 cycles. Tumor tissues were taken from patients before they received treatment with anti-CD40 Ab.1. For example, samples were taken as part of routine lymphoma diagnosis.
Clinical Trial 002 (Phase I)
Multi-institutional, multi-dose phase I study was conducted to test the safety, pharmacokinetic properties, immunogenicity, and antitumor activity of intravenous anti-CD40 Ab.1 in patients with relapsed NHL. Patients with multiple histologic subtypes of NHL were enrolled on this study, including diffuse large B-cell (DLBCL; 14), follicular (FCL; 9), mantle cell (MCL; 9), marginal zone (MZL; 2) and small lymphocytic (SLL; 1). Patients were treated with a dose-loading schedule: 1 mg/kg of anti-CD40 Ab.1 on day 1 and day 4 and subsequent intra-patient dose-escalation during weeks 2-5 to a maximum dose of 3, 4, 6, or 8 mg/kg over four cohorts. Subsequently, a rapid dose-loading schedule was tested in one cohort (40% increase in total anti-CD40 Ab.1 administered during cycle 1). Responding patients or those with stable disease were eligible for a second cycle, consisting of four consecutive weekly infusions at the cohort-specific maximum dose of anti-CD40 Ab.1. Eight patients with DLBCL completed cycle 1 and received a maximum dose of at least 3 mg/kg anti-CD40 Ab.1 with an objective response rate of 37.5% (i.e. 1 CR and 2 PR) and 2 SD. Additional objective responses were seen in one patient with MCL (CR) and one patient with MZL (PR). The median duration of response for these 5 patients has not yet been reached (range 8-37 weeks). Tumor tissues were taken from patients before they received treatment with anti-Cd40 Ab.1. For example, samples were taken as part of routine lymphoma diagnosis.
Clinical Sample Preparation and qRT-PCR
Formalin Fixed Paraffin Embedded (FFPE) archival tumor tissue from the Phase I and Phase II clinical trials described above was obtained from the clinical investigation sites with appropriate IRB approval and patient consent. 4-6 micron sections derived from the tumor tissue were mounted on glass slides and one slide for each case was subject to H&E staining using standard pathology laboratory protocol. A board certified Pathologist marked the H&E slide for tumor content and was used as a guide to macrodissect the remaining tumor-containing region for RNA extraction using the Ambion RecoverAll™ Total Nucleic Acid Isolation Kit for FFPE Tissues (Cat. No. AM1975; Applied Biosystems/Ambion, Austin, Tex.).
450 ng total RNA per sample was reverse transcribed in a total reaction volume of 20 uL using Applied Biosystems' High Capacity Reverse Transcription cDNA Synthesis kit (Cat. No. 4368814; Applied Biosystems, Foster City, Calif.). Manufacturer's recommendations were followed with the exception of a shortened 60 min RT reaction at 37 degrees. 5 ng total RNA equivalent cDNA (assuming 100% cDNA synthesis efficiency) product was mixed with Applied Biosystems' 2× Universal Master Mix (no UNG) in a volume of 15 uL for each PCR assay well. All amplifications were performed in triplicate in 384-well plates using a 2-step (95 degrees 15 sec, 60 degrees 1 min) PCR amplification procedure. Reactions were carried out to 40 cycles on a validated ABI 7900 real-time PCR system. Sequences of the primers and probes used are shown in Table 10.
TABLE 10
Primers and Probes
Probe
Gene GenBank Over-
Locus Accession No. lap Forward Primer Reverse Primer Probe
PRKCA NM_002737.2 1 TGACAAAATGTAGAGGCCATTCA CATCCGTCTCCTCTGCGATATAA CCGTCAAACACCATTT
(SEQ ID NO: 3) (SEQ ID NO: 4) (SEQ ID NO: 5)
IGF1R NM_000875.3 1 TTGCAAGGAAAGAAATTCAAACAC TGCTTGAATCCATTGACTGCTT ACAACAGCAGTAAGAAGA
(SEQ ID NO: 6) (SEQ ID NO: 7) (SEQ ID NO: 8)
BTG2 NM_006763.2 1 CAGGTCCCTGCCTTTTTAGAAG ATCATAAAGAAGAGAAGAGAGACA  AGCCTCATGGTCTCAT
(SEQ ID NO: 9) GAATTAAG  (SEQ ID NO: 11)
(SEQ ID NO: 10)
LMO2 NM_005574.2 1 GGCCACAGCCCATCCA CTTGCCCCTAAATGTTCCTTTCT AGTAACTGACATGATTAGC
(SEQ ID NO: 12) (SEQ ID NO: 13) (SEQ ID NO: 14)
CD22 NM_001771.2 1 TTTGGAAGTGAGGCATTGCA CCGGAGTCCCCAGAGTCAA AGACGTACGTATCAGCG
(SEQ ID NO: 15) (SEQ ID NO: 16) (SEQ ID NO: 17)
SMN1 NM_000344.2 1 CTGGAATGTGAAGCGTTATAGAAG CCTTTTTTCTTTCCCAACACTTGA CTGGCCTCATTTCT
AT (SEQ ID NO: 19) (SEQ ID NO: 20)
(SEQ ID NO: 18)
EPDR1 NM_017549.3 1 CAGCCTCTCTTGTCCCTGGTT TCCCTAGCAATGGACAAACTCA CCTTATGTGTTGAATGTGG
(SEQ ID NO: 21) (SEQ ID NO: 22) (SEQ ID NO: 23)
CD40 NM_001250.4 1 GGGATCCTGTTTGCCATCCT GCTTCTTGGCCACCTTTTTG TTGGTGCTGGTCTTT
(SEQ ID NO: 24) (SEQ ID NO: 25) (SEQ ID NO: 26)
IFITM1 NM_003641.3 1 GGCTTCATAGCATTCGCCTACT TCACGTCGCCAACCATCTT CGTGAAGTCTAGGGACAG
(SEQ ID NO: 27) (SEQ ID NO: 28) (SEQ ID NO: 29)
VNN2 NM_004665.2 1 GACTTGTATGTATGGGAGTGAGGA TCTCTTCAAGGGCACAGCTATG CAGGGCCATTGCAA
GTT (SEQ ID NO: 31) (SEQ ID NO: 32)
(SEQ ID NO: 30)
PRPSAP NM_002767.2 1 GCCAAACTGGAAACATAAGAGTGA GCATGACGGTTCCTGTGAAA TGCTCGGTGGGATGG
2 (SEQ ID NO: 33) (SEQ ID NO: 34) (SEQ ID NO: 35)
PRKCA NM_002737.2 1 CGGAGGTTGAGGTTTTTCCTT GACGGTTGAATGGCCTCTACA TGTATAAGCACCTACTGACA
(SEQ ID NO: 36) (SEQ ID NO: 37) AA (SEQ ID NO: 38)
IGF1R NM_000875.3 1 AGGACTTCTTCATGGGTCTTACAG AAGTGACATTAAAGACGATGTGTA TGTTAGACCATGAAACATT
TT TGC (SEQ ID NO: 41)
(SEQ ID NO: 39) (SEQ ID NO: 40)
BTG2 NM_006763.2 1 CAGGCTGTGTTCTTGCATCTTG GACCATGAGGCTGCTTCTAAAAA CTGCAAACAGGTCCCT
(SEQ ID NO: 42) (SEQ ID NO: 43) (SEQ ID NO: 44)
LMO2 NM_005574.2 1 TTGGACCCAAGGGAAAACTG GGTTAAAAGTTGTGGTTTCCATTC TGGAGACGCATTTCG
(SEQ ID NO: 45) TC (SEQ ID NO: 47)
(SEQ ID NO: 46)
CD22 NM_001771.2 1 GACATCCCCACTCACGAATATTAT CTGTCCTTTTCTGGGCTTTCC CCAGTTTCTGCCTCTGA
G (SEQ ID NO: 49) (SEQ ID NO: 50)
(SEQ ID NO: 48)
SMN1 NM_000344.2 1 GGCATAGAGCAGCACTAAATGACA TTCTATAACGCTTCACATTCCAGA CACTAAAGAAACGATCAGAC
(SEQ ID NO: 51) TC (SEQ ID NO: 53)
(SEQ ID NO: 52)
EPDR1 NM_017549.3 0 CGCACTTTGGCCTTCCTAGA TGGAAGGAGATGCAGAAGTCAGA CACTGCTTCATAACCTC
(SEQ ID NO: 54) (SEQ ID NO: 55) (SEQ ID NO: 56)
CD40 NM_001250.4 1 CCTGCCCAGTCGGCTTCT GTCCAAGGGTGACATTTTTCG CTCCAATGTGTCATCTG
(SEQ ID NO: 57) (SEQ ID NO: 58) (SEQ ID NO: 59)
IFITM1 NM_003641.3 1 GGGTTACTAGTAGCCGCCCATA GCAGGGCCAGCATTGC CAACCTTTGCACTCCAC
(SEQ ID NO: 60) (SEQ ID NO: 61) (SEQ ID NO: 62)
VNN2 NM_004665.2 1 TGTCCATTTTTTTGGCTACTCTGA CCCAAACACCCAGGCTCTT CAGTGTGGAACAATG
(SEQ ID NO: 63) (SEQ ID NO: 64) (SEQ ID NO: 65)
PRPSAP NM_002767.2 0 GCTCCAGTGCCCCAAGATT CGACGGATCGCCTCTGAA AAACTGTGGATATCAGCATG
2 (SEQ ID NO: 66) (SEQ ID NO: 67) A 
(SEQ ID NO: 68)
PRKCA NM_002737.2 0 TGGGCAACTCAGAAATACTTCGA ACGTCAATAGGCACGTTTGCT CTCCCAAGATATAAGAGGC
(SEQ ID NO: 69) (SEQ ID NO: 70) (SEQ ID NO: 71)
IGF1R NM_000875.3 0 GTCCACCCTCTCCCCTTTCT CACGCACTCTAGTACAAAGCATAA CTCACTCCAAGAAAC
(SEQ ID NO: 72) GA (SEQ ID NO: 74)
(SEQ ID NO: 73)
BTG2 NM_006763.2 0 CCCAAACCGAATCACCTTAAGA CAGGAGGGTGGCCATCCT ACAGGGCTAGGGCAT
(SEQ ID NO: 75) (SEQ ID NO: 76) (SEQ ID NO: 77)
LMO2 NM_005574.2 0 TCTCCATGGCATCTTCGTCTT ATCCCTTACCCCACCCTCAA ACTCTTAGGCACTTTGG
(SEQ ID NO: 78) (SEQ ID NO: 79) (SEQ ID NO: 80)
CD22 NM_001771.2 0 CGGCCTCAGGCACAAGAA GCAGCCCATCCAGTGTCAAT ATGTGGACTATGTGATCCT
(SEQ ID NO: 81) (SEQ ID NO: 82) (SEQ ID NO: 83)
SMN1 NM_000344.2 0 CATGGTACATGAGTGGCTATCATA GTGAGCACCTTCCTTCTTTTTGA CTATTATATGGGTTTCAGAC
CTG (SEQ ID NO: 85) AAA
(SEQ ID NO: 84) (SEQ ID NO: 86)
EPDR1 NM_017549.3 0 GACTATTGTCTCCTAAACCCAGGA CCCAGTGCATTTAATGACCAAA AGTTCCCTCGTACTGTC
CTA (SEQ ID NO: 88) (SEQ ID NO: 89)
(SEQ ID NO: 87)
CD40 NM_001250.4 1 ATCAATTTTCCCGACGATCTTC CGGTTGGCATCCATGTAAAGT TGGCTCCAACACTG
(SEQ ID NO: 90) (SEQ ID NO: 91) (SEQ ID NO: 92)
IFITM1 NM_003641.3 0 AGGTCCACCGTGATCAACATC CAGGGACCAGACGACATGGT ACAGCGAGACCTCCGT
(SEQ ID NO: 93) (SEQ ID NO: 94) (SEQ ID NO: 95)
VNN2 NM_004665.2 0 CAACTTGTGGACGGCCAGTA GTGCCACTGAGGGAGAACATTT AAACTGCTTCTACAAGATT
(SEQ ID NO: 96) (SEQ ID NO: 97) (SEQ ID NO: 98)
PRPSAP NM_002767.2 0 CAGCAGAGACCCTGAAGGAAA CAAGCCATGAGTTGCCATCA AGGTGCATATAAGATCTT
2 (SEQ ID NO: 99) (SEQ ID NO: 100) (SEQ ID NO: 101)
BCL6 NM_001706.2 1 CCCATTCTGCGTCATGCTT AATGCAGTTTAGACACAGCCAAAC TGTTATAACTACTCCGGAGA
(SEQ ID NO: 102) (SEQ ID NO: 103) CAG(SEQ ID NO: 104)
LRRC8A NM_019594.2 1 AGTTCAGCCCAGATGGAAGGT GCGGCATCGCTAAATAAGGA TTCAGGGAAAGGTGGGC
(SEQ ID NO: 105) (SEQ ID NO: 106) (SEQ ID NO: 107)
BCL6 NM_001706.2 1 CACAGGGACTTGAAGTTGTTACTA TGACGCAGAATGGGATGAGA CTCTCTTTGGGAATGTT
ACTAA (SEQ ID NO: 109) (SEQ ID NO: 110)
(SEQ ID NO: 108)
LRRC8A NM_019594.2 0 CAAAGCAGCCAGACGTTGAAC CACACCAGATCCGGAAGACA TTTCCCTGGGCGCAGG
(SEQ ID NO: 111) (SEQ ID NO: 112) (SEQ ID NO: 113)
RGS13 NM_144766.1 0 GGGATTCCTACCCCAGATTTCTA CAGAAACTGTTGTTGGACTGCATAG AGTCAGAAATGTACCAAAAA
(SEQ ID NO: 114) (SEQ ID NO: 115) (SEQ ID NO: 116)
YIPF3 NM_015388.2 1 TGAGCTGTAGCTGCGTAAGTACCT GGCCTTGTGCCTTTCAGAAG CTTGATGCCTGTCGGC
(SEQ ID NO: 117) (SEQ ID NO: 118) (SEQ ID NO: 119)
YIPF3 NM_015388.2 1 TGGCTGCCCTACACATGCT CAGGATCCCCTCTACCACTTTG CCTGCTCTATCTGCATTT
(SEQ ID NO: 120) (SEQ ID NO: 121) (SEQ ID NO: 122)
YIPF3 NM_015388.2 0 GAGGCTCAGCTGTGATTGACAT CACCCATATCCTCGAAGCTAGAG AGAACATGGATGATACCTC
(SEQ ID NO: 123) (SEQ ID NO: 124) (SEQ ID NO: 125)
RGS13 NM_44766.1 0 TCCAGCCACAGTCCCCTAGA TCCTGAATGTTCCTGATGATAGTCT AGATTAACATTGACAGTTCG
(SEQ ID NO: 126) CT ACA(SEQ ID NO: 128)
(SEQ ID NO: 127)
EPDR1 NM_017549.3 0 CGAGAGGAAGGCGCTGATC ACATCACTCCATCCTTATACAGCAA CCTGCAAGAGATTATTT
(SEQ ID NO: 129) A (SEQ ID NO: 131)
(SEQ ID NO: 130)
EPDR1 NM_017549.3 0 GGATCCTCTTGACATTCCTCAAA GGCCCCCCGATGGA CTCCACCTTTGAAGACC
(SEQ ID NO: 132) (SEQ ID NO: 133) (SEQ ID NO: 134)
EPDR1 NM_017549.3 0 CGAGGGTGTGGCCATATGA GAACAGGCATTAGAAATACCCAAAG TGACTAGATGGCTAATATG
(SEQ ID NO: 135) (SEQ ID NO: 136) (SEQ ID NO: 137)
UAP1 NM_003115.4 0 CTACTGCAAGGCATGCTTTGAT TGGCCCCCTGCATTGA TCCCTTCATCATTGCTG
(SEQ ID NO: 138) (SEQ ID NO: 139) (SEQ ID NO: 140)
CD79B NM_000626.2 0 GCCGGTGCAGTTACACGTT CCCCAAACCCGTGACAAC CCTCCAAGGAGCCTC
(SEQ ID NO: 141) (SEQ ID NO: 142) (SEQ ID NO: 143)
CLPTM1 NM_001294.1 1 CAAGGCCCTCAACACATTCA GGTACATAACGGGCATCTTGATG ACCTGTTCGCCTTTG
(SEQ ID NO: 144) (SEQ ID NO: 145) (SEQ ID NO: 146)
UAP1 NM_003115.4 1 CCTATGCTGGAGAAGGATTAGAAA CGATGATTAGAGGTGCATGGAA ATGTGGCAGATAAAG
GT (SEQ ID NO: 148) (SEQ ID NO: 149)
(SEQ ID NO: 147)
CD79B NM_000626.2 0 TCTCGCCACCCTCACCAT GCTGACAGAAGTAGATGCCATTGT CAAGGCATCCGGTTTG
(SEQ ID NO: 150) (SEQ ID NO: 151) (SEQ ID NO: 152)
CLPTM1 NM_001294.1 0 AAGTCGCCCTGGAACTTCCT CACCGAGTCCTGCTCCTCAT ATGAGTTGTACGAGCAGTC
(SEQ ID NO: 153) (SEQ ID NO: 154) (SEQ ID NO: 155)
UAP1 NM_003115.4 1 CATGAGCTGGTGAAAAATGGTAT AAAGCTATTCCTATCGTGGCAAA AACCAGATACCAAGTTTT
TT (SEQ ID NO: 157) (SEQ ID NO: 158)
(SEQ ID NO: 156)
CD79B NM_000626.2 1 TCCCCAGCTCTTGCCAAAG CAGAGAACTCCCTCCAAGTTGCT CTGGAGTAGAAGGACAACAG
(SEQ ID NO: 159) (SEQ ID NO: 160) (SEQ ID NO: 161)
CLPTM1 NM_001294.1 0 GGCAGGCCAGGGTTTGT CGAGATGGCTGGAAACACAGA AGGCGCTGTCTGTC
(SEQ ID NO: 162) (SEQ ID NO: 163) (SEQ ID NO: 164)
CTSC NM_001814.3 1 GACTCAGCCTCTGGGATGGA GGATCCGGAAGTAGCCATTCT TGGATTGTTAAAAACAGCTG
(SEQ ID NO: 165) (SEQ ID NO: 166) G (SEQ ID NO: 167)
CTSC NM_001814.3 0 AGGCGGCTTCCCATACCT CTTCTTCCACCAGCCCAAAA ATTGCAGGAAAGTACGCC
(SEQ ID NO: 168) (SEQ ID NO: 169) (SEQ ID NO: 170)
CTSC NM_001814.3 0 CCCAAACCTGCACCACTGA CAAGATGTTGGCAAATGCAAA CTGAAATACAGCAAAAGA
(SEQ ID NO: 171) (SEQ ID NO: 172) (SEQ ID NO: 173)
CD44 NM_000610.3 0 CCTTTGTGGCATTTATTCATCAGT GCTTCTATGACAAGCAGCCTTTG AGGGTGTCCGATTGG
(SEQ ID NO: 174) (SEQ ID NO: 175) (SEQ ID NO: 176)
PUS7 NM_019042.3 0 CTCTGTAGCACAGGCTGGATTG AGGCTGCAGTGCAAGATTGA AGTGCAATCCTGCAATT
(SEQ ID NO: 177) (SEQ ID NO: 178) (SEQ ID NO: 179)
CD44 NM_000610.3 0 CCACTTGGAGGCCTTTCATC AGGTTGGCGATCAGGAATACA TCGGGTGTGCTATGGA
(SEQ ID NO: 180) (SEQ ID NO: 181) (SEQ ID NO: 182)
PUS7 NM_019042.3 0 CCTTGCCTGGTTTCGATGTT GAGCATTTCCCTGTAGGCTTCTT CCCAAAGCATAAAATT
(SEQ ID NO: 183) (SEQ ID NO: 184) (SEQ ID NO: 185)
CD44 NM_000610.3 0 CAACCGTTGGAAACATAACCATT AACAATCAGTAGCACATTGCATCTG AGGGAGCTGGGACACT
(SEQ ID NO: 186) (SEQ ID NO: 187) (SEQ ID NO: 188)
PUS7 NM_019042.3 0 TGGACTCACTGAGGCTGACGTA GATTCCCGAGAACCCTTGATG TCACCAAGTTTGTGAGTTC
(SEQ ID NO: 189) (SEQ ID NO: 190) (SEQ ID NO: 191)
RPL22 NM_000983.3 1 GCTGCCAATTTTGAGCAGTTT GTTCCCAGCTTTTCCGTTCA TGCAAGAAAGGATCAAA
(SEQ ID NO: 192) (SEQ ID NO: 193) (SEQ ID NO: 194)
LOC728 XR_015348.1 1 TCTTGCCTGCCCTGTGTTG TGCCTTCCCCTTAATAATGCA AAAATGCGGGTCCCTT
179 (SEQ ID NO: 195) (SEQ ID NO: 196) (SEQ ID NO: 197)
SERBP1 NM_001018067.1 1 CTCCCGCTACACAGAAGTAACAAA AAAACATCCCTGCTACCAATACATT ATGGTAGTCAGTTTTGTATT
(SEQ ID NO: 198) (SEQ ID NO: 199) TAG(SEQ ID NO: 200)
RPL9 NM_000661.4 1 TCCGTTACAAGATGAGGTCTGTGT CATTCTCCTGGATAACAACGTTGA TGCTCACTTCCCC
(SEQ ID NO: 201) (SEQ ID NO: 202) (SEQ ID NO: 203)
CFL1 NM_005507.2 1 TCCATCCCTTGACGGTTCTG AGCCCAAGAGGAATCAAAAGATC CCTTCCCAAACTGCTTT
(SEQ ID NO: 204) (SEQ ID NO: 205) (SEQ ID NO: 206)
RPL13 NM_000977.2 1 GAGTCATCACTGAGGAAGAGAAGA TGGCACGGGCCATACG CAAAGCCTTCGCTAGTC
ATT (SEQ ID NO: 208) (SEQ ID NO: 209)
(SEQ ID NO: 207)
FLJ160 NM_198505.1 1 CCTACACCCCTTATCCCCATACT CCAGGGCTATTGGTTGAATGA TTATTATCGAAACCATCAGC
25 (SEQ ID NO: 210) (SEQ ID NO: 211) C 
(SEQ ID NO: 212)
RPS10 NM_001014.3 1 CGACCTGCGAGACTCACAAG GGCACAGCACTCCGTCTGT AAGCTGACAGAGATACC
(SEQ ID NO: 213) (SEQ ID NO: 214) (SEQ ID NO: 215)
NPM1 NM_002520.5 1 TCTGGCTGTCCTTTTTATAATGCA CTTGGCAATAGAACCTGGACAAC AGTGAGAACTTTCCC
(SEQ ID NO: 216) (SEQ ID NO: 217) (SEQ ID NO: 218)
CCDC72 NM_015933.3 1 GCAAGAAGAAGCCACTGAAACA GAAAGCCTTATCTTCCTCGTCCAT CCCAAGAAGCAGGCCA
(SEQ ID NO: 219) (SEQ ID NO: 220) (SEQ ID NO: 221)
RPS19 NM_001022.3 1 GGCTGAAAATGGTGGAAAAGG CTTTGTCCCTGAGGTGTCAGTTT CCAAGATGGCGGCCG
(SEQ ID NO: 222) (SEQ ID NO: 223) (SEQ ID NO: 224)
RPS16 NM_001020.4 1 TGTGGATGAGGCTTCCAAGAA CAGCAGGGTCCGGTCATACT AGATCAAAGACATCCTCATC
(SEQ ID NO: 225) (SEQ ID NO: 226) (SEQ ID NO: 227)
EEF1G NM_001404.4 1 GGCAGGTGGACTACGAGTCATAC GTCTCCTCGCTGCCAGGAT CATGGCGGAAACTG
(SEQ ID NO: 228) (SEQ ID NO: 229) (SEQ ID NO: 230)
RPS5 NM_001009.3 1 CCGGAACATTAAGACCATTGC CCCTTGGCAGCATTGATGA AGTGCCTGGCAGATG
(SEQ ID NO: 231) (SEQ ID NO: 232) (SEQ ID NO: 233)
EEF1A1 NM_001402.5 1 CTGCCACCCCACTCTTAATCA GGCCAATTGAAACAAACAGTTCT TGGTGGAAGAACGGTC
(SEQ ID NO: 234) (SEQ ID NO: 235) (SEQ ID NO: 236)
RPL28 NM_000991.3 1 GGAAGCCTGCCACCTCCTAT TGGCGCGAGCATTCTTG TGCGGACCACCATC
(SEQ ID NO: 237) (SEQ ID NO: 238) (SEQ ID NO: 239)
ACTG1 NM_001614.2 1 TGTCCTTGAAGCTTGTATCTGATA TTCAATACAAGGTCAAAATCAGCAA CACTGGATTGTAGAACTT
TCA (SEQ ID NO: 241) (SEQ ID NO: 242)
(SEQ ID NO: 240)
BTF3 NM_001037637.1 1 AGCCTCAGATGAAAGAAACAATCA CACTTGTGCCTGCAGTTTGG AACCAGGAAAAACTC
(SEQ ID NO: 243) (SEQ ID NO: 244) (SEQ ID NO: 245)
TMSB4X NM_021109.2 1 AAGCAGGCGAATCGTAATGAG TGCTTGTGGAATGTACAGTGCAT CGTGCGCCGCCAA
(SEQ ID NO: 246) (SEQ ID NO: 247) (SEQ ID NO: 248)
TPM3 NM_153649.3 1 CCCTTTTCTGGGTTTGAAGCT CTGACTGATACAAAGCACAATTGAG CTGTCTCTAGAAGTGCC
(SEQ ID NO: 249) A (SEQ ID NO: 251)
(SEQ ID NO: 250)
USMG5 NM_032747.2 1 GCTGTGAAAGCAACATAAATGGAT GGCATGGGAACTTAACAGATGAG TTAAACTGTCTACGGTTCTT
(SEQ ID NO: 252) (SEQ ID NO: 253) (SEQ ID NO: 254)
EIF1 NM_005801.3 1 CGCTATCCAGAACCTCCACTCT CAGGTCATCACCCTTACTTGCA TCGACCCCTTTGCTG
(SEQ ID NO: 255) (SEQ ID NO: 256) (SEQ ID NO: 257)
Data Processing
The raw qRT-PCR as results were pre-processed according to the description below under Normalization, Transformation, and Imputation and the Sensitivity Index was computed as described under Sensitivity Index and Classifier. Spearman's rank correlations were used for correlation estimates and corresponding P-values. For the Multivariate Sensitivity Index, probes were selected and coefficients estimated using the elastic net blend of lasso (L1) and ridge (L2) penalized regression, as described by Zhou et al., Statist. Soc. B. 67:301-320, 2005 and implemented by Friedman, Hastie and Tibshirani, Regularization Paths for Generalized Linear Models via Coordinate Descent. Technical Report, Dept. of Statistics, Stanford University at www-stat.stanford.edu/˜hastie/Papers/glmnet.pdf. X2 tests were used to test for associations among categorical variables.
Normalization, Transformation and Imputation
The following are definitions for assay data and model parameters:
Definitions
Assay Data
    • l=a reference set of samples (e.g. NHL cell lines)
    • Nl=sample size
    • p=number of probes (not including normalizers)
    • Nlj (Obs)=detected sample size for probe j
    • Nlj (ND)=not detected sample size for probe j
    • yij (Obs)=detected raw assay value for sample i, probe j
    • pi (nrm.Obs)=number of detected normalizer values for sample i
    • yij (nrm.Obs)=detected normalizer value for sample i, probe j
Model Parameters
    • {circumflex over (μ)}lj (Obs.raw)=set l mean of detected log2 assay values for probe j (un-normalized)
    • {circumflex over (σ)}lj (Obs)=set l standard deviation of detected log2 assay values for probe j
    • γl (ND)=set l number of standard deviations above the mean
      For a reference set of samples, such as that used to fit index coefficients and classifier cutoffs, mean and standard deviation model parameters are computed using the reference set data (refer to the formulas for Reference Set Model Parameters below). For new samples, for example a single new sample for which the index and class are to be computed, model parameters must be taken from a reference set, l, which is chosen to be the most representative of the population from which the new sample is drawn. For example, a clinical reference set for each indication and line of therapy in which the assay is used may be maintained. The formulas for calculating reference set model parameters and transformed, normalized assay values are shown below.
Formulas
Reference Set Model Parameters
Intermediate Values
μ ^ i ( nrm , Obs ) = 1 p i ( nrm , Obs ) j = 1 p i ( nrm , Obs ) y ij ( nrm , Obs ) ( sample normalization factor ) μ ^ lj ( Obs ) = 1 N lj ( Obs ) i = 1 N lj ( Obs ) [ log 2 ( y ij ( Obs ) ) - log 2 ( μ ^ i ( nrm , Obs ) ) ] ( normalized mean )
Model Parameters
σ ^ lj ( Obs ) = 1 N lj ( Obs ) i = 1 N lj ( Obs ) ( log 2 ( y ij ( Obs ) ) - log 2 ( μ ^ i ( nrm , Obs ) ) - μ ^ lj ( Obs ) ) 2 μ ^ lj ( Obs , raw ) = 1 N lj ( Obs ) i = 1 N lj ( Obs ) log 2 ( y ij ( Obs ) )
Transformed, Normalized Assay Values
Intermediate Values
μ ^ i ( nrm , Obs ) = 1 p i ( nrm , Obs ) j = 1 p i ( nrm , Obs ) y ij ( nrm , Obs ) ( sample normalization factor )
Transformed, Normalized, Imputed Assay Values
x ij (Obs)=−[log2(y ij (Obs))−log2({circumflex over (μ)}i (nrm.Obs))], i=1, . . . , N lj (ND)
x ij (N D)=−[{circumflex over (μ)}lj (Obs.raw)−log2({circumflex over (μ)}i (nrm.Obs))+γl (N D){circumflex over (σ)}lj (Obs) ], i=1, . . . , N lj (ND)
    • The completed Nl×p matrix of values,
[ x 1 ( Obs ) x p ( Obs ) x 1 ( ND ) x p ( ND ) ] ,
is input to the sensitivity index and classifier calculations.
Sensitivity Index and Classifier
The following are definitions for assay data and model parameters:
Definitions
Assay Data
    • l=a reference set of samples (e.g. NHL cell lines)
    • Nl=sample size
    • p=number of probe pairs
    • xij=transformed, normalized assay value for sample i, probe j
    • xij′=as above with j′ the anti-correlated pair probe to probe j
Model Parameters
    • βlj=set l coefficient for probe j
    • {circumflex over (μ)}lj=set l mean of transformed normalized assay values for probe j
    • {circumflex over (σ)}lj 2=set l mean of transformed normalized assay values for probe j
    • Ll=classification cutpoint
      The formulas for calculating reference set model parameters and sensitivity index and classifier are shown below.
Formulas
Reference Set Model Parameters
Probe Means and Standard Deviations μ ^ lj = 1 N l i = 1 N l x ij σ ^ lj 2 = 1 N l i = 1 N l ( x ij - μ ^ lj ) 2
Index and Classifier
Sensitivity Index S li = j = 1 p β lj x ij - μ ^ lj σ ^ lj 2 - β lj x ij - μ ^ lj σ ^ lj 2 Sensitivity Class T li = { 1 sensitive if S li C l 0 resistant otherwise
Clinical Trial 001 Results
Table 11 below provides a sample accounting of assayed specimens and clinical samples from Clinical Trial 001. Twenty nine archival FFPE tumor specimens from 24 patients with DLBCL were submitted for qRT-PCR processing. Three patients had multiple specimens and all 24 patients had usable qRT-PCR results for at least one specimen. Of these 24, 21 had tumor sum of the product of diameters (SPD) measurements reported both at baseline and at least one post-baseline visit.
TABLE 11
Clinical Trial 001 Sample Accounting
Diagnostic Assay Clinical Database
Archival FFPE 29 Analysis
specimens sample size
# of patients (3 24 (both qRT-PCR and
with multiple SPD available)
specimens)
Specimens 27
qRT-PCR
Reported
Usable qRT-PCR 26 46 Patients in clinical
results database
(1 insufficient)
qRT-PCR for 24 21 39 SPD Change from
unique patients Baseline Reported
(2 patient
specimen pairs
averaged
together)
Table 12 summarizes the pairwise Spearman's rank correlations between the Main and Pair genes that contribute to the sensitivity index. Based on the cell line development samples, genes with low expression in particular groups of patient should be expected to have relatively high expression of the corresponding pair, on average, providing for self-normalization and the interpretation of the Sensitivity Index as a ratio of up- to down-regulated expression pathways (i.e. on a log base 2 scale). The magnitude of the correlations between pairs in this first clinical sample are statistically significant and notable high throughout, with the lower correlation estimate being −0.67 (P=0.0004). These tests alone constitute an independent confirmation that the assay target sequences are expressed in tumor samples from this clinical population in-vitro and that the assay is detecting expression in the archived FFPE tissue samples.
TABLE 12
Main and Pair Gene Anti-correlations (N = 21)
Main Locus Correlation
Gene* Link Gene Pair
IFITM1 8519 −.85 BTG2
CD40 958 −.84 IGF1R
RGS13 6003 −.70 CD44
VNN2 8875 −.87 CTSC
LMO2 4005 −.67 EPDR1
CD79B 974 −.75 UAP1
CD22 933 −.83 PUS7
*CD40, RGS13, VNN2, LMO2, CD22, BTG2, and UAP1 are genes with higher expression in sensitive cell lines.
Table 13 summarizes the associations between the measurements for each probe individually and the largest reduction (or smallest increase) in tumor SPD post-baseline. Since rank correlations are based upon the difference (or ratio) of post-baseline to baseline measurements, positive correlations mean that higher expression of the probe is associated with tumor increases, on average; and the negative correlations mean that higher expression of the probe is associated with tumor decreases on average. Notably, all Main-Pair probe pairs have opposite-direction associations with SPD. The P-values are consistent with a promising trend in this sample. All P-values are below 0.5 (50% expected when there is no true association). All ranges are calculated as bootstrap 95th percentile confidence intervals, based upon 5,000 replicates sampled with replacement from the DLBCL patient sample, N=21. Narrower ranges will become available as the sample size increases. Since no model-building or checking was required to produce these results, they comprise a robust trend, which confirms that these qRT-PCR probe measurements are associated, overall, with reduction in tumor SPD in patients treated with anti-CD40 Ab. 1.
TABLE 13
Associations between SPD and Individual Probe Measurements (N = 21)
Main Pair
Gene Rho. P Range Gene Rho. P Range
IFITM1 +0.29 0.20 (−0.13, 0.68) BTG2 −0.27 0.23 (−0.70, 0.19)
CD40 −0.16 0.49 (−0.58, 0.30) IGF1R +0.33 0.15 (−0.17, 0.73)
RGS13 −0.32 0.16 (−0.66, 0.13) CD44 +0.34 0.14 (−0.11, 0.70)
VNN2 −0.26 0.26 (−0.67, 0.21) CTSC +0.31 0.17 (−0.17, 0.68)
LMO2 −0.25 0.27 (−0.69, 0.25) EPDR1 +0.27 0.23 (−0.22, 0.67)
CD79B +0.22 0.34 (−0.22, 0.61) UAP1 −0.22 0.35 (−0.59, 0.22)
CD22 −0.25 0.28 (−0.66, 0.21) PUS7 +0.20 0.39 (−0.26, 0.66)
The multivariate sensitivity index is a weighted average of the probes in Tables 12 and 13. Since weights in cell lines were not expected to reflect optimal weights in patient tumor specimens, the weights in cell lines were restricted to 1 and −1, corresponding to the signed, equal-weighted average, where the signs matched the association between each probe and resistance to anti-CD40 Ab.1 by IC25 in the cell lines. For clinical populations, new weights are required. As a preliminary analysis based upon 21 samples only, we chose to use a penalized, multivariate regression procedure to select and estimate weights for the best 8 of the 14 probes. Those weights (coefficient) are shown in Table 14, and the association between the resulting Sensitivity Index and SPD change from baseline is depicted in FIG. 7. Larger multivariate Sensitivity Index values are associated with SPD decreases post-baseline (Spearman's Rho=−0.58, P=0.006). All ranges in Tables 13, 14, and 15 were calculated as bootstrap 95th percentile confidence intervals, based upon 5,000 replicates sampled with replacement from the DLBCL patient sample, N=21. Narrower ranges will become available as the sample size increases.
TABLE 14
Weights for the Multivariate Sensitivity Index (N = 21)
Main Pair
Gene Coeff. Range Gene Coeff. Range
IFITM1 −0.08 (−11.7, 3.7) BTG2 −0.62 (−11.6, 0.0)
CD40 0 (−9.5, 8.2) IGF1R 0 (−9.0, 5.6)
RGS13 +1.13 (−1.9, 8.0) CD44 −3.39 (−11.9, 0.0)
VNN2 0 (−4.1, 4.1) CTSC 0 (−8.8, 2.1)
LMO2 0 (−8.5, 2.1) EPDR1 −0.74 (−4.7, 3.6)
CD79B +0.04 (−3.2, 9.0) UAP1 −2.45 (−15.1, 0.0)
CD22 +0.63 (−0.0, 12.7) PUS7 0 (−7.7, 7.3)
Using 26 samples from Clinical Trail 001, ranges for μj and σj values obtained are as shown in Table 15.
TABLE 15
μj and σj ranges based on data from Clinical Trail 001
μj IFITM1 LMO2 CD40 VNN2 IGF1R BTG2 CD22 BCL6
lower −4.89 −5.09 −5.09 −5.10 −5.12 −5.02 −5.03 −5.07
upper −4.79 −5.00 −5.02 −5.02 −5.06 −4.92 −4.93 −4.99
μj RGS13 EPDR1 CD79B UAP1 CTSC CD44 PUS7
lower −5.14 −5.19 −5.10 −5.26 −5.04 −4.97 −5.24
upper −5.00 −5.12 −5.04 −5.18 −4.95 −4.87 −5.16
σj IFITM1 LMO2 CD40 VNN2 IGF1R BTG2 CD22 BCL6
lower 0.10 0.09 0.07 0.08 0.06 0.09 0.09 0.08
upper 0.17 0.14 0.12 0.13 0.10 0.15 0.14 0.12
σj RGS13 EPDR1 CD79B UAP1 CTSC CD44 PUS7
lower 0.14 0.07 0.06 0.08 0.09 0.09 0.08
upper 0.22 0.11 0.10 0.12 0.14 0.16 0.12
Clinical Trial 002 Results
Raw qRT-PCR results were successfully generated for 10 patients with archival specimens. For those 10 patients, diagnosis, treatment group, multivariate sensitivity index, clinical response and SPD change from baseline are shown in Table 16. The multivariate sensitivity index weights were taken from the 21 Clinical Trial 001 patients (Table 14), so that these patients constitute a very small validation set. 2 of 4 patients with Sensitivity Index ≧0 exhibited some tumor shrinkage after anti-CD40 Ab.1 exposure and 4 of 6 patients with Sensitivity Index <0 exhibited either tumor increase or a best response of PD (SPD was unavailable for 2 patients, but a best clinical response outcome was available for this patient).
TABLE 16
Summary of diagnosis, treatment group, multivariate sensitivity index,
clinical response and SPD change for 6 patients in Clinical Trial 002.
Treatment Sensitivity Best SPD Percent
Samples Dx. Group Index Response Change
066-0001 MCL Pre-2 +0.01 PD +72.48
066-0015 MCL V −0.87 PD +64.07
066-0009 DLBCL III +1.06 PR −78.02
066-0006 DLBCL I −2.31 PR −66.44
066-0011 T-Cell- IV −0.46 SD (PR) −10.34
LBCL
066-0005 DLBCL I −2.99 PD +1,208.94
066-0013 MCL IV −3.67 PD +94.59
066-0019 DLBCL V +0.15 SD −32.64
066-0004 DLBCL I −0.46 PD ?
066-0002 DLBCL Pre-2 +0.99 PD ?
BCL6. The qRT-PCR assay contains a 15th probe for the BCL6 gene. Though not currently used in the multivariate Sensitivity Index, it was a previously identified potential predictor of response to anti-CD40 Ab.1. As shown in FIG. 8, while not significantly associated with SPD change in the combined DLBCL patient sample (P=0.25, N=26), BCL6 trends lower in those with tumor increases (rho=−0.23).
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention.

Claims (16)

What is claimed is:
1. A method for treating a human subject having a B-cell lymphoma, comprising the steps of:
(a) performing a nucleic acid-based detection assay to detect the expression level of the UAP1 RNA transcript in a sample comprising B lymphoma cells obtained from the human subject;
(b) determining that the B lymphoma cells from the human subject express the UAP1 RNA transcript at an increased level compared to a reference level;
(c) determining that the human subject is responsive to an anti-CD40 antibody treatment based on the increased level of the UAP1 RNA transcript in the B lymphoma cells as compared to the reference level; and
(d) administering an effective amount of an anti-CD40 antibody to the human subject expressing the increased level of the UAP1 RNA transcript in the B lymphoma cells, thereby treating B-cell lymphoma in the human subject.
2. The method of claim 1, wherein the expression level of the UAP1 RNA transcript is normalized.
3. The method of claim 1, wherein the reference level is determined based on the expression level of the UAP1 RNA transcript in samples comprising B lymphoma cells from subjects having tumor volume decreased after the anti-CD40 antibody treatment.
4. The method of claim 3, wherein the samples from subjects for reference level determination comprise the same type of B lymphoma cells as the sample from the subject whose responsiveness to the anti-CD40 antibody treatment is determined.
5. The method of claim 1, wherein the anti-CD40 antibody stimulates CD40 and enhances the interaction between CD40 and CD40 ligand.
6. The method of claim 5, wherein the anti-CD40 antibody comprises the heavy chain amino acid sequence shown in SEQ ID NO:1 and the light chain amino acid sequence shown in SEQ ID NO:2.
7. The method of claim 1, wherein the anti-CD40 antibody stimulates CD40 and does not enhance or inhibits the interaction between CD40 and CD40 ligand.
8. The method of claim 1, further comprising performing a nucleic acid-based detection assay to detect the expression levels of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, or at least thirteen marker genes in addition to UAP1, wherein the marker genes are selected from the group consisting of IFITM1, CD40, RGS13, VNN2, LMO2, CD79B, CD22, BTG2, IGF1R, CD44, CTSC, EPDR1, and PUS7.
9. The method of claim 8, wherein the expression levels of IFITM1, RGS13, CD79B, CD22, BTG2, CD44, EPDR1, and UAP1 are detected.
10. The method of claim 1, wherein the B cell lymphoma is diffuse large B-cell lymphoma (DLBCL).
11. The method of claim 1, wherein the B cell lymphoma is non-Hodgkin's lymphoma.
12. The method of claim 11, wherein the non-Hodgkin's lymphoma is follicular lymphoma, mantle cell lymphoma, marginal zone lymphoma, or small lymphocytic lymphoma.
13. The method of claim 1, wherein the sample comprising the B lymphoma cells is a formalin fixed paraffin embedded biopsy sample.
14. The method of claim 1, wherein the RNA transcript is measured by qRT-PCR.
15. The method of claim 1, further comprising assaying the expression level of BCL6, wherein an increased expression of BCL6 as compared to a reference level determines that the subject is responsive to the anti-CD40 antibody treatment.
16. The method of claim 15, wherein the reference level is determined based on the expression level of BCL6 in samples comprising B lymphoma cells from subjects having tumor volume decreased after the anti-CD40 antibody treatment.
US12/741,814 2007-11-07 2008-11-07 Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies Active US9068230B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/741,814 US9068230B2 (en) 2007-11-07 2008-11-07 Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US98627707P 2007-11-07 2007-11-07
PCT/US2008/082920 WO2009062125A1 (en) 2007-11-07 2008-11-07 Methods and compositions for assessing responsiveness of b-cell lymphoma to treatment with anti-cd40 antibodies
US12/741,814 US9068230B2 (en) 2007-11-07 2008-11-07 Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies

Publications (2)

Publication Number Publication Date
US20110104671A1 US20110104671A1 (en) 2011-05-05
US9068230B2 true US9068230B2 (en) 2015-06-30

Family

ID=40377361

Family Applications (1)

Application Number Title Priority Date Filing Date
US12/741,814 Active US9068230B2 (en) 2007-11-07 2008-11-07 Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies

Country Status (11)

Country Link
US (1) US9068230B2 (en)
EP (1) EP2222875B1 (en)
JP (2) JP5730576B2 (en)
KR (1) KR101620642B1 (en)
CN (1) CN101910414B (en)
AU (1) AU2008323701B2 (en)
BR (1) BRPI0820707A2 (en)
CA (1) CA2704499C (en)
IL (1) IL205391A (en)
MX (1) MX2010005080A (en)
WO (1) WO2009062125A1 (en)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009131365A2 (en) * 2008-04-21 2009-10-29 한국생명공학연구원 Cst1, dcc1, ifitm1 or melk as markers for diagnosing stomach cancer
US9365901B2 (en) 2008-11-07 2016-06-14 Adaptive Biotechnologies Corp. Monitoring immunoglobulin heavy chain evolution in B-cell acute lymphoblastic leukemia
US9528160B2 (en) 2008-11-07 2016-12-27 Adaptive Biotechnolgies Corp. Rare clonotypes and uses thereof
US8628927B2 (en) 2008-11-07 2014-01-14 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
CN102272327B (en) 2008-11-07 2015-11-25 赛昆塔公司 By the method for sequential analysis monitoring situation
US9506119B2 (en) 2008-11-07 2016-11-29 Adaptive Biotechnologies Corp. Method of sequence determination using sequence tags
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
DK2387627T3 (en) 2009-01-15 2016-07-04 Adaptive Biotechnologies Corp Adaptive immunity profiling and methods for producing monoclonal antibodies
ES2605228T3 (en) * 2009-04-18 2017-03-13 Genentech, Inc. Methods to assess the responsiveness of a B-cell lymphoma to treatment with anti-CD40 antibodies
KR20120044941A (en) 2009-06-25 2012-05-08 프레드 헛친슨 켄서 리서치 센터 Method of measuring adaptive immunity
EP2580348B1 (en) * 2010-06-14 2018-04-25 Qiagen GmbH Method for determination of target cells or tissue for extraction of biomolecules from non-formalin-fixed biological samples
JP6271254B2 (en) * 2011-02-28 2018-01-31 ジェネンテック, インコーポレイテッド Methods for predicting biological markers and responses to B cell antagonists
US8592156B2 (en) * 2011-08-08 2013-11-26 Roche Molecular Systems, Inc. Predicting response to anti-CD20 therapy in DLBCL patients
US20140356339A1 (en) * 2011-09-09 2014-12-04 Sequenta Inc. Sequence-based measures of immune response
US10385475B2 (en) 2011-09-12 2019-08-20 Adaptive Biotechnologies Corp. Random array sequencing of low-complexity libraries
AU2012325791B2 (en) 2011-10-21 2018-04-05 Adaptive Biotechnologies Corporation Quantification of adaptive immune cell genomes in a complex mixture of cells
EP3904536A1 (en) 2011-12-09 2021-11-03 Adaptive Biotechnologies Corporation Diagnosis of lymphoid malignancies and minimal residual disease detection
US9499865B2 (en) 2011-12-13 2016-11-22 Adaptive Biotechnologies Corp. Detection and measurement of tissue-infiltrating lymphocytes
US10077478B2 (en) 2012-03-05 2018-09-18 Adaptive Biotechnologies Corp. Determining paired immune receptor chains from frequency matched subunits
CN107586832B (en) 2012-05-08 2021-03-30 适应生物技术公司 Compositions and methods for measuring and calibrating amplification bias in multiplex PCR reactions
AU2013327423B2 (en) 2012-10-01 2017-06-22 Adaptive Biotechnologies Corporation Immunocompetence assessment by adaptive immune receptor diversity and clonality characterization
WO2015160439A2 (en) 2014-04-17 2015-10-22 Adaptive Biotechnologies Corporation Quantification of adaptive immune cell genomes in a complex mixture of cells
US9708657B2 (en) 2013-07-01 2017-07-18 Adaptive Biotechnologies Corp. Method for generating clonotype profiles using sequence tags
ES2738289T3 (en) * 2013-11-06 2020-01-21 Hospital Clinic Barcelona Procedure for the subtyping of lymphoma types by means of expression profiles
EP3114240B1 (en) 2014-03-05 2019-07-24 Adaptive Biotechnologies Corporation Methods using randomer-containing synthetic molecules
RU2016136716A (en) * 2014-03-27 2018-04-28 Дженентек, Инк. METHODS FOR DIAGNOSTIC AND TREATMENT OF INFLAMMATORY DISEASES OF THE GUT
US10066265B2 (en) 2014-04-01 2018-09-04 Adaptive Biotechnologies Corp. Determining antigen-specific t-cells
EP3715455A1 (en) 2014-10-29 2020-09-30 Adaptive Biotechnologies Corp. Highly-multiplexed simultaneous detection of nucleic acids encoding paired adaptive immune receptor heterodimers from many samples
JP6691113B2 (en) 2014-10-29 2020-04-28 シアトル ジェネティックス, インコーポレイテッド Dosage and administration of non-fucosylated anti-CD40 antibody
US10246701B2 (en) 2014-11-14 2019-04-02 Adaptive Biotechnologies Corp. Multiplexed digital quantitation of rearranged lymphoid receptors in a complex mixture
US11066705B2 (en) 2014-11-25 2021-07-20 Adaptive Biotechnologies Corporation Characterization of adaptive immune response to vaccination or infection using immune repertoire sequencing
CA2976580A1 (en) 2015-02-24 2016-09-01 Adaptive Biotechnologies Corp. Methods for diagnosing infectious disease and determining hla status using immune repertoire sequencing
EP3277294A4 (en) 2015-04-01 2018-11-14 Adaptive Biotechnologies Corp. Method of identifying human compatible t cell receptors specific for an antigenic target
EP3359688B1 (en) 2015-10-05 2021-06-16 UCB Biopharma SRL Molecular signatures for use in diagnosis and response to treatment analysis of autoimmune diseases
CN109414451B (en) * 2016-05-16 2022-08-12 德克萨斯大学系统董事会 Compositions for delivering tRNA as nanoparticles and methods of use thereof
IL263834B2 (en) 2016-06-20 2024-01-01 Kymab Ltd Anti-pd-l1 antibodies
US10428325B1 (en) 2016-09-21 2019-10-01 Adaptive Biotechnologies Corporation Identification of antigen-specific B cell receptors
CN110402255B (en) * 2017-04-04 2022-12-02 豪夫迈·罗氏有限公司 Novel bispecific antigen binding molecules capable of specifically binding to CD40 and FAP
US11254980B1 (en) 2017-11-29 2022-02-22 Adaptive Biotechnologies Corporation Methods of profiling targeted polynucleotides while mitigating sequencing depth requirements

Citations (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4016043A (en) 1975-09-04 1977-04-05 Akzona Incorporated Enzymatic immunological method for the determination of antigens and antibodies
US4018653A (en) 1971-10-29 1977-04-19 U.S. Packaging Corporation Instrument for the detection of Neisseria gonorrhoeae without culture
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4424279A (en) 1982-08-12 1984-01-03 Quidel Rapid plunger immunoassay method and apparatus
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
US5182368A (en) 1986-06-13 1993-01-26 Ledbetter Jeffrey A Ligands and methods for augmenting B-cell proliferation
US5445934A (en) 1989-06-07 1995-08-29 Affymax Technologies N.V. Array of oligonucleotides on a solid substrate
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
WO1996018413A1 (en) 1994-12-13 1996-06-20 The University Of Birmingham Carcinoma treatment
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5591669A (en) 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
WO1997017852A1 (en) 1995-11-15 1997-05-22 Hoechst Schering Agrevo Gmbh Synergetic herbicidal mixtures
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
WO1997031025A1 (en) 1996-02-23 1997-08-28 Chiron Corporation Humanized anti-cd40 monoclonal antibodies and fragments capable of blocking b cell activation
US5674492A (en) 1993-12-23 1997-10-07 Immunex Corporation Method of preventing or treating disease characterized by neoplastic cells expressing CD40
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO2000075348A1 (en) 1999-06-08 2000-12-14 Seattle Genetics, Inc. Recombinant anti-cd40 antibody and uses thereof
US6248535B1 (en) 1999-12-20 2001-06-19 University Of Southern California Method for isolation of RNA from formalin-fixed paraffin-embedded tissue specimens
WO2001075166A2 (en) 2000-03-31 2001-10-11 Genentech, Inc. Compositions and methods for detecting and quantifying gene expression
WO2001083755A2 (en) 2000-04-28 2001-11-08 La Jolla Institute For Allergy And Immunology Human anti-cd40 antibodies and methods of making and using same
US20020197256A1 (en) 2001-04-02 2002-12-26 Genentech, Inc. Combination therapy
WO2003040170A2 (en) 2001-11-09 2003-05-15 Pfizer Products Inc. Antibodies to cd40
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2004071572A2 (en) 2003-02-06 2004-08-26 Genomic Health, Inc. Gene expression markers for response to egfr inhibitor drugs
US20050090434A1 (en) * 2003-06-13 2005-04-28 Morris David W. Novel therapeutic targets in cancer
WO2005044294A2 (en) 2003-11-04 2005-05-19 Chiron Corporation Method of therapy for cancers expressing the cd40 antigen
US20050164231A1 (en) * 2003-09-03 2005-07-28 Staudt Louis M. Methods for identifying, diagnosing, and predicting survival of lymphomas
US20050260646A1 (en) 2004-04-09 2005-11-24 Genomic Health Inc. Gene expression markers for predicting response to chemotherapy
WO2006125117A2 (en) 2005-05-18 2006-11-23 Novartis Ag Methods for diagnosis and treatment of diseases having an autoimmune and/or inflammatory component
WO2006125143A2 (en) 2005-05-18 2006-11-23 Novartis Ag Methods for diagnosis and treatment of proliferative disorders mediated by cd40 signaling
WO2006128103A2 (en) 2005-05-26 2006-11-30 Seattle Genetics, Inc. Humanized anti-cd40 antibodies and their methods of use
WO2006133420A2 (en) 2005-06-08 2006-12-14 Millennium Pharmaceuticals, Inc. Treatment of patients with cancer therapy
JP2006342173A (en) 2001-04-27 2006-12-21 Kirin Brewery Co Ltd Anti-cd40 monoclonal antibody
WO2007032743A2 (en) 2005-09-16 2007-03-22 Biovator Technologies Ab In vitro assay for identification of allergenic proteins
US20070110754A1 (en) 2003-11-04 2007-05-17 Chiron Corporation Use of antagonist anti-cd40 antibodies for treatment of chronic lymphocytic leukemia
WO2007066230A2 (en) 2005-10-31 2007-06-14 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method for identifying, marking and treating epithelial lung tumour cells and means for carrying out said method
WO2007075348A1 (en) 2005-12-22 2007-07-05 Basf Catalysts Llc Muffler assembly having a metal catalyst therein
WO2007082379A2 (en) 2006-01-20 2007-07-26 Mcgill University Method to identify cd40-sensitive cells using gene expression
WO2008079269A2 (en) 2006-12-19 2008-07-03 Genego, Inc. Novel methods for functional analysis of high-throughput experimental data and gene groups identified therfrom
WO2010121231A1 (en) 2009-04-18 2010-10-21 Genentech, Inc. Methods for assessing responsiveness of b-cell lymphoma to treatment with anti-cd40 antibodies

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006044294A2 (en) 2004-10-14 2006-04-27 Eli Lilly And Company Human protein c analogs

Patent Citations (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4018653A (en) 1971-10-29 1977-04-19 U.S. Packaging Corporation Instrument for the detection of Neisseria gonorrhoeae without culture
US4016043A (en) 1975-09-04 1977-04-05 Akzona Incorporated Enzymatic immunological method for the determination of antigens and antibodies
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4424279A (en) 1982-08-12 1984-01-03 Quidel Rapid plunger immunoassay method and apparatus
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US5182368A (en) 1986-06-13 1993-01-26 Ledbetter Jeffrey A Ligands and methods for augmenting B-cell proliferation
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5591669A (en) 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US5445934A (en) 1989-06-07 1995-08-29 Affymax Technologies N.V. Array of oligonucleotides on a solid substrate
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5674492A (en) 1993-12-23 1997-10-07 Immunex Corporation Method of preventing or treating disease characterized by neoplastic cells expressing CD40
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
WO1996018413A1 (en) 1994-12-13 1996-06-20 The University Of Birmingham Carcinoma treatment
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1997017852A1 (en) 1995-11-15 1997-05-22 Hoechst Schering Agrevo Gmbh Synergetic herbicidal mixtures
WO1997031025A1 (en) 1996-02-23 1997-08-28 Chiron Corporation Humanized anti-cd40 monoclonal antibodies and fragments capable of blocking b cell activation
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2000075348A1 (en) 1999-06-08 2000-12-14 Seattle Genetics, Inc. Recombinant anti-cd40 antibody and uses thereof
US6946129B1 (en) 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
US6843989B1 (en) 1999-06-08 2005-01-18 Seattle Genetics, Inc. Methods for the treatment and prevention of cancer using anti-CD40 antibodies
US6838261B1 (en) 1999-06-08 2005-01-04 Seattle Genetics, Inc. Nucleic acids encoding anti-CD40 proteins and methods of producing recombinant anti-CD40 proteins
US6248535B1 (en) 1999-12-20 2001-06-19 University Of Southern California Method for isolation of RNA from formalin-fixed paraffin-embedded tissue specimens
WO2001075166A2 (en) 2000-03-31 2001-10-11 Genentech, Inc. Compositions and methods for detecting and quantifying gene expression
WO2001083755A2 (en) 2000-04-28 2001-11-08 La Jolla Institute For Allergy And Immunology Human anti-cd40 antibodies and methods of making and using same
US20020197256A1 (en) 2001-04-02 2002-12-26 Genentech, Inc. Combination therapy
JP2006342173A (en) 2001-04-27 2006-12-21 Kirin Brewery Co Ltd Anti-cd40 monoclonal antibody
WO2003040170A2 (en) 2001-11-09 2003-05-15 Pfizer Products Inc. Antibodies to cd40
CN1582165A (en) 2001-11-09 2005-02-16 辉瑞产品公司 Antibodies to CD40
US7288251B2 (en) 2001-11-09 2007-10-30 Abgenix, Inc. Antibodies to CD40
WO2004071572A2 (en) 2003-02-06 2004-08-26 Genomic Health, Inc. Gene expression markers for response to egfr inhibitor drugs
US20050090434A1 (en) * 2003-06-13 2005-04-28 Morris David W. Novel therapeutic targets in cancer
US20050164231A1 (en) * 2003-09-03 2005-07-28 Staudt Louis M. Methods for identifying, diagnosing, and predicting survival of lymphomas
WO2005044294A2 (en) 2003-11-04 2005-05-19 Chiron Corporation Method of therapy for cancers expressing the cd40 antigen
US20070110754A1 (en) 2003-11-04 2007-05-17 Chiron Corporation Use of antagonist anti-cd40 antibodies for treatment of chronic lymphocytic leukemia
US20050260646A1 (en) 2004-04-09 2005-11-24 Genomic Health Inc. Gene expression markers for predicting response to chemotherapy
WO2006125117A2 (en) 2005-05-18 2006-11-23 Novartis Ag Methods for diagnosis and treatment of diseases having an autoimmune and/or inflammatory component
WO2006125143A2 (en) 2005-05-18 2006-11-23 Novartis Ag Methods for diagnosis and treatment of proliferative disorders mediated by cd40 signaling
JP2008539794A (en) 2005-05-18 2008-11-20 ノバルティス アーゲー Methods for the diagnosis and treatment of proliferative disorders mediated by CD40 signaling
WO2006128103A2 (en) 2005-05-26 2006-11-30 Seattle Genetics, Inc. Humanized anti-cd40 antibodies and their methods of use
WO2006133420A2 (en) 2005-06-08 2006-12-14 Millennium Pharmaceuticals, Inc. Treatment of patients with cancer therapy
JP2008544223A (en) 2005-06-08 2008-12-04 ミレニアム・ファーマシューティカルズ・インコーポレイテッド Methods for identifying, determining and treating patients undergoing cancer therapy
WO2007032743A2 (en) 2005-09-16 2007-03-22 Biovator Technologies Ab In vitro assay for identification of allergenic proteins
WO2007066230A2 (en) 2005-10-31 2007-06-14 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method for identifying, marking and treating epithelial lung tumour cells and means for carrying out said method
WO2007075348A1 (en) 2005-12-22 2007-07-05 Basf Catalysts Llc Muffler assembly having a metal catalyst therein
WO2007082379A2 (en) 2006-01-20 2007-07-26 Mcgill University Method to identify cd40-sensitive cells using gene expression
WO2008079269A2 (en) 2006-12-19 2008-07-03 Genego, Inc. Novel methods for functional analysis of high-throughput experimental data and gene groups identified therfrom
WO2010121231A1 (en) 2009-04-18 2010-10-21 Genentech, Inc. Methods for assessing responsiveness of b-cell lymphoma to treatment with anti-cd40 antibodies
US20120123695A1 (en) 2009-04-18 2012-05-17 David Dornan Methods for assessing responsiveness of b-cell lymphoma to treatment with anti-cd40 antibodies

Non-Patent Citations (140)

* Cited by examiner, † Cited by third party
Title
Advani, R. et al. (2009). "Evaluation of a Gene Signature to Predict Single Agent Dactuzumab (SGN-40) Activity in Patients with DLBCL," Abstract No. 11063 presented at 2009 ASCO Annual Meeting, three pages, located at <http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst-detail-view&confID=65&abstractIDS=32462>, last visited on Aug. 2, 2010, J Clin Oncol 27:15S.
Anonoymous. (2011). "The R Project for Statistical Computing," located at , last visited on Feb. 18, 2011, one page.
Anonoymous. (2011). "The R Project for Statistical Computing," located at <http://www.r-project.org/main.shtml>, last visited on Feb. 18, 2011, one page.
Anonoymous. (Nov. 7, 2003). "Affymetrix GenoChip Human Genome U133 Plus 2.0 Array," Geo Expression, two pages.
Ausubel et al. (1995). Short Protocols in Molecular Biology, Current Protocols in Molecular Biology, pp. iii-xvii. (Table of Contents Only).
Ausubel, F.M. (1987). Current Protocols in Molecular Biology, New York, New York, five pages. (Table of Contents Only.).
Barbas, C.F. et al. (Apr. 26, 1994). "In Vitro Evolution of a Neutralizing Human Antibody to Human Immunodeficiency Virus Type 1 to Enchance Affinity and Broaden Strain cross-Reactivity," Proc Nat. Acad. Sci USA 91(9):3809-3813.
Basso, K. et al. (Dec. 15, 2004, e-pub. Aug. 26, 2004). "Tracking CD40 Signaling During Germinal Center Development," Blood 104:4088-4096.
Bohen, S.P. et al. (Feb. 18, 2004). "Variation in Gene Expression Patterns in Follicular Lymphoma and the Response to Rituximab," Proceedings of the National Academy of Sciences of the United States of American 100(4):1926-1930.
Bruggemann, M. et al. (1993). "Designer Mice:the Production of Human Antibody Repertoires in Transgenic Animals," Year in Immuno. 7:33-40.
Canellos G.P. et al. (1998). The Lymphomas. W.B.Saunders Company, Philadelphia, Pennsylvania, pp. xi-xiii. (Table of Contents Only.).
Cheung, V.G. et al. (Jan. 1999). "Making and Reading Microarrays," Nature Genetics 21(Suppl):15-19.
Clackson, T. et al. (Aug. 15, 1991). "Making Antibody Fragments Using Phage Display Libraries," Nature 352(6336):624-628.
Clynes, R. et al. (Jan. 20, 1998). "Fc Receptors are Required in Passive and Active Immunity to Melanoma," PNAS USA 95(2):652-656.
Coleman, R. Drug Discovery Today. 2003. 8: 233-235. *
Coligen et al. (1991). Current Protocols in Immunology, vols. 1 and 2, Ed. Wiley-Interscience, New York, New York, Pubs, one page. (Table of Contents Only.).
Definition for "Assay", printed on Dec. 1, 2014, available via url: . *
Definition for "Assay", printed on Dec. 1, 2014, available via url: <https://www.google.com/search?q=assaying+definition>. *
Dermer, G.B. Bio/Technology (1994) 12: 320. *
Dogan et al. American Journal Surgical Pathology. 2000. 24(6): 846-852. *
Dogan, A. et al. (2000). "CD10 and BCL-6 Expression in Paraffin Sections of Normal Lymphoid Tissue and B-Cell Lymphomas," Amercian Journal of Surgical Pathology 24(6):846-852.
Dornan, D. et al. (Nov. 20, 2009). "CD40 Pathway Activation Status Predicts Response to CD40 Targeted Therapy in Diffuse Large b-Cell Lymhoma," 51st Annual Meeting of the American Society of Hematology, New Orleans, Lousiana, Dec. 5 through 8, 2009, four pages, Blood 114(22):1065.
Fellouse, F.A. (Aug. 24, 2004, e-pub. Aug. 11, 2004). "Synthetic Antibodies From a Four-Amino-Acid Code: A Dominant Role for Tyrosine in Antigen Recognition," Proc. Nat. Acad. Sci. USA 101(34):1246712472.
Fishwild, D.M et al., (Jul. 1996). "High-Avidity Human IgG Kappa Monoclonal Antibodies from a Novel Strain of Minilocus Transgentic Mice," Nature Biotechnol 14(7):845-851.
Forero-Torres et al. (2006). "A humanized antibody against CD40 (SGN-40) is well tolerated and active in non-Hodgkin's lymphoma (NHL): Results of phase I Study," Journal of Clinical Oncology 24(18S): Abstract No. 7534, one page.
Forero-Torres, A. et al. (Nov. 20, 2009). "A Phase 1b Clinical Trial of Dacetuzumab in Combination with Rituximab and Gemcitabine: Multiple Responses Observed in Patients with Relasped Diffuse Large B-Cell Lymphomia," 51st Annual Meeting of the American Society of Hematology, New Orleans, Lousiana, Dec. 5 though 9, 2009, four pages. Blood 114(22):243-244.
Francisco, J.A. et al. (Jun. 15, 2000). "Agonistic Properties and In Vivo Antitumor Activity of the Anti-CD40 Antibody SGN-14," Cancer Res. 60(12):3225-3231.
Freshney, R.I. ed. (1987). Animal Cell Culture, Oxford, England, pp. ix-xiv. (Table of Contents.).
Friedman J.et al. (2008). "Regularization Paths for Generalized Linear Models via Coordinate Descent." Technical Report, Department of Statistics, Stanford University (World Wide Webstat.stanford.edui-hastie/Papers/glmnet.pdf) R package glmnet; R Development Core Team.
Gait, M.J. ed. (1984). Oligonucleotide Synthesis, Oxford, England, pp. vii-xii. (Table of Contents Only.).
GenBank Accession No. AY143166, last updated Dec. 5, 2003, located at , last visited May 15, 2011, two pages.
GenBank Accession No. AY143166, last updated Dec. 5, 2003, located at <http://www.ncbi.nlm.nih.gov/nuccore/AY143166>, last visited May 15, 2011, two pages.
GenBank Accession No. BC019297, last updated Jul. 15, 2006, located at , last visited May 15, 2011, one page.
GenBank Accession No. BC019297, last updated Jul. 15, 2006, located at <http://www.ncbi.nlm.nih.gov/nuccore/BC019297>, last visited May 15, 2011, one page.
GenBank Accession No. BC062723, last updated Sep. 1, 2006, located at , last visited May 15, 2011, three pages.
GenBank Accession No. BC062723, last updated Sep. 1, 2006, located at <http://www.ncbi.nlm.nih.gov/nuccore/BC062723>, last visited May 15, 2011, three pages.
GenBank Accession No. NM-000344, last updated May 1, 2011, located at , last visited May 15, 2011, five pages.
GenBank Accession No. NM-000344, last updated May 1, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-000344>, last visited May 15, 2011, five pages.
GenBank Accession No. NM-000610.3, last updated Aug. 5, 2012, located at , last visited Aug. 31, 2012, eight pages.
GenBank Accession No. NM-000610.3, last updated Aug. 5, 2012, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-000610>, last visited Aug. 31, 2012, eight pages.
GenBank Accession No. NM-000626, last updated on Jun. 26, 2012, located at , last visited Aug. 31, 2012, four pages.
GenBank Accession No. NM-000626, last updated on Jun. 26, 2012, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-000626>, last visited Aug. 31, 2012, four pages.
GenBank Accession No. NM-000875, last updated May 15, 2011, located at , last visited May 15, 2011, one page.
GenBank Accession No. NM-000875, last updated May 15, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-000875>, last visited May 15, 2011, one page.
GenBank Accession No. NM001250, last updated May 14, 2011, located at , last visited May 15, 2011, three pages.
GenBank Accession No. NM001250, last updated May 14, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/nm-001250>, last visited May 15, 2011, three pages.
GenBank Accession No. NM-001706, last updated Apr. 30, 2011, located at , last visited May 15, 2011, three pages.
GenBank Accession No. NM-001706, last updated Apr. 30, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-001706>, last visited May 15, 2011, three pages.
GenBank Accession No. NM-001771, last updated Mar. 13, 2011, located at , last visited May 15, 2011, four pages.
GenBank Accession No. NM-001771, last updated Mar. 13, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-001771>, last visited May 15, 2011, four pages.
GenBank Accession No. NM-001814.4, last updated Apr. 6, 2008, located at, NCBI, NLM (Bethesda, MD).
GenBank Accession No. NM-002737, last updated Oct. 7, 2008, located at , last visited May 15, 2011, seven pages.
GenBank Accession No. NM-002737, last updated Oct. 7, 2008, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-002737>, last visited May 15, 2011, seven pages.
GenBank Accession No. NM-002767, last updated Mar. 11, 2011, located at , last visited May 15, 2011, one page.
GenBank Accession No. NM-002767, last updated Mar. 11, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-002767>, last visited May 15, 2011, one page.
GenBank Accession No. NM-002927, last updated Mar. 20, 2011, located at , last visited May 15, 2011, four pages.
GenBank Accession No. NM-002927, last updated Mar. 20, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-002927>, last visited May 15, 2011, four pages.
GenBank Accession No. NM-003115.4, last updated Jun. 27, 2012, located at , last visited on Aug. 31, 2012, four pages.
GenBank Accession No. NM-003115.4, last updated Jun. 27, 2012, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-003115>, last visited on Aug. 31, 2012, four pages.
GenBank Accession No. NM-003641, last updated May 14, 2011, located at , last visited May 15, 2011, one page.
GenBank Accession No. NM-003641, last updated May 14, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/AJ132100>, last visited May 15, 2011, one page.
GenBank Accession No. NM-004665, last updated Mar. 11, 2011, located at , last visited May 15, 2011, one page.
GenBank Accession No. NM-004665, last updated Mar. 11, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-004665>, last visited May 15, 2011, one page.
GenBank Accession No. NM-005574, last updated Apr. 10, 2011, located at , last visited May 15, 2011, three pages.
GenBank Accession No. NM-005574, last updated Apr. 10, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-005574>, last visited May 15, 2011, three pages.
GenBank Accession No. NM-006763 last updated Mar. 13, 2011, located at , last visited May 15, 2011, three pages.
GenBank Accession No. NM-006763 last updated Mar. 13, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-006763>, last visited May 15, 2011, three pages.
GenBank Accession No. NM-015388, last updated Jul. 21, 2012, located at , last visited Sep. 6, 2012, three pages.
GenBank Accession No. NM-015388, last updated Jul. 21, 2012, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-015388 >, last visited Sep. 6, 2012, three pages.
GenBank Accession No. NM-015883, last updated Oct. 6, 2003, located at , last visited Jun. 13, 2011, one page.
GenBank Accession No. NM-015883, last updated Oct. 6, 2003, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-015883.1?report=genbank>, last visited Jun. 13, 2011, one page.
GenBank Accession No. NM-017549, last updated Mar. 11, 2011, located at , last visited May 15, 2011, three pages.
GenBank Accession No. NM-017549, last updated Mar. 11, 2011, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-017549>, last visited May 15, 2011, three pages.
GenBank Accession No. NM-019042, last updated Feb. 11, 2008, located at , last visited Aug. 31, 2012, three pages.
GenBank Accession No. NM-019042, last updated Feb. 11, 2008, located at <http://www.ncbi.nlm.nih.gov/nuccore/NM-019042>, last visited Aug. 31, 2012, three pages.
GeneAnnot for BCL6 (available via url: <genecards.weizmann.ac.il/cgi-bin/geneannot/GA-search.pl?array=HG-U133&array=HG-U133-Plus'2&keyword-type=gene-symbol&keyword=bcl6&target=integrated&.submit=Submit+Query>, printed on Mar. 28, 2013). *
GeneCard for BCL6 (available via url: , printed on Mar. 28, 2013). *
GeneCard for BCL6 (available via url: < genecards.org/cgi-bin/carddisp.pl?gene=BCL6>, printed on Mar. 28, 2013). *
GeneCard for UAP1, available via url: <genecards.org/cgi-bin/carddisp.pl?gene=UAP1> printed on Oct. 3, 2012. *
GeneCard for UAP1, available via url: printed on Oct. 3, 2012. *
Gokmen-Polar et al. Cancer Research. 2001. 61: 1375-1381. *
Hammerling, G. J. et al. (1981). "Monoclonal Antibodies and T-Cell Hybridomas." Chapter 3 in Production of Antibody-Producing Hybridomas in the Rodent Systems, Elsevier/North-Holland Biomedical Press, Amsterdam, Holland, pp. 563-681.
Harlow, E. et al. (1988). Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, pp. iii-ix. (Table of Contents Only.).
Hawkins, R.E. et al. (Aug. 5, 1992). "Selection of Phage Antibodies by Binding Affinity. Mimicking Affinity Maturation," I Mot Biol. 226(3):889-896.
Haynes et al Electrophoresis. 1998. 19: 1862-1871. *
International Search Report mailed on Aug. 24, 2010, for PCT Application No. PCT/US2010/031528, filed on Apr. 17, 2010, four pages.
International Search Report mailed on Mar. 11, 2009, for PCT Application No. PCT/US2008/082920, filed on Nov. 7, 2008, three pages.
Jackson, J.R. et al. (Apr. 1995). "In Vitro Antibody Maturation Improvements of High Affinity Neutralizing Antibody Againist IL-1 Beta," J. Immunol. 154(7):3310-9.
Jakobovits, A. et al. (1993). "Analysis of Homozygous Mutant Chimeric Mice: Deletion of the Immunoglobulin Heavy-Chain Joining Region Blocks B-Cell Development and Antibody Production," Proc. Natl. Acad. Sci. USA 90(6):2551-2555.
Jakobovits, A. et al. (Mar. 18, 1993). "Germ-Line Transmission and Expression of A human-Derived Yeast Artificial Chrmosome," Nature 362(6417):255-258.
Jones, P.T. et al. (May 29-Jun. 4, 1986). "Replacing the Complementarity-Determing Regions in a Human Antibody with Those from a Mouse," Nature 321(6069):522-525.
Kimura, H. et al. (Jun. 2005, e-pub. Sep. 8, 2005). "Plasma MIP-1beta levels and skin toxicity in Japanese non-small cell lung cancer patients treated with the EGFR-targeted tyrosine kinase inhibitor, gefitinib," Lung Cancer 50(3):393-399.
Kimura, H. et al. (Jun. 2005, e-pub. Sep. 8, 2005). "Plasma MIP-1β levels and skin toxicity in Japanese non-small cell lung cancer patients treated with the EGFR-targeted tyrosine kinase inhibitor, gefitinib," Lung Cancer 50(3):393-399.
Kohler, G. et al. (Aug. 7, 1975). "Continuous Cultures of Fused Cells Secreting Antibody of Predefined Specifcity," Nature 256(5517):495-497.
Lee, C.V. et al. (Jan. 2004). "Bivalent Antibody Phage Display Mimics Natural Immunoglbulin," J. Immunol. Methods 284(1-2):119-132.
Lee, C.V. et al. (Jul. 23, 2004). "High-Affinity Human Antibodies From Phage-Displayed Synthetic Fab Libraries with a Single Framework Scaffold," J.Mol.Biol. 340(5):1073-1093.
Leong, A.S.Y. et al. (1996). "Epitope Retrieval with Microwaves. A Comparison of Citrate Buffer and EDTA with Tree Commercial Retrieval Solutions," Appl. Immunohistochem. 4(3):201-207.
Liu et al . Clinical Immunology. 2004. 112: 225-230. *
Lockhart, D.J. (Dec. 1996). "Expression Monitoring by Hybridization to High-Density Oligonucleotide Arrays," Nature Biotechnology 14(13):1675-1680.
Lonberg, N. et al. (1995). "Human Antibodies From Transgenic Mice," Intern. Rev. Immunol. 13:65-93.
Lonberg, N. et al. (Apr. 28, 1994). "Antigen-Specific Human Antibodies From Mice Comprising Distinct Genetic Modifications," Nature 368(6474):856-859.
Lossos, I.S. et al. (Aug. 15, 2001). "Expression of a Single Gene, BCL-6, Strongly Predicts Survival in Patients With Diffuse Large B-Cell Lymphona," 98(4):945-951.
Luna, L.G. (1960). "Manual of Histological Staining Method of the Armed Forces Institute of Pathology," 3rd edition, The Blakston Division McGraw-Hill Book Company, New York, New York.
Marks, J.D. et al. (Dec. 5, 1991). "By-Passing Immunization. Human Antibodies From V-Gene Libraries Displayed on Phage," J. Mol. Biol. 222(3):581-597.
Marks, J.D. et al. (Jul. 1992). "By-Passing Immunization: Building High Affinity Human Antibodies by Chain Shuffling," Biotechnology 10(7):779-783.
Mikel, U.V. eds. (1994). "The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology," Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C.
Morrison, S.L. (Apr. 28, 1994). "Immunology. Success in Specification," Nature 368:(6474)812-813.
Morrison, S.L. et al. (Nov. 1984). "Chimeric Human Antibody Molecules: Mouse Antigen-Binding Domains with Human Constant Region Domains," Proc. Natl. Acad. Sci. USA 81(21):6851-6855.
Mullis et al. eds. (1994). "PCR: The Polymerase Chain Reaction" Birkäuser, Boston, MA, pp. xvi-xvii. (Table of Contents Only.).
Neuberger, M. (Jul. 1996). "Generating High-Advidity Human Mabs in Mice," Nature Biotechnol. 14(7):826.
Nutt et al. (Apr. 1, 2003). "Gene expression-based classification of malignant gliomas correlates better with survival than histological classification," Cancer Research 63:1602-1607.
O'Sullivan et al. (1981). "Methods for the Preparation of Enzyme-Antibody Conjugates for use in Enzyme Immunoassay," Chap. 73 in Methods in Enzymology, J. Langone et al eds., Academic Press, New York, New York, pp. 147-166.
Overbergh, L. et al. (Mar. 2003). "The Use of Real-Time Reverse Transcriptase PCR for the Quantification of Cytokine Gene Expression," J. Biomolecular Techniques 14(1):33-43.
Presta, L.G. (Aug. 1992). "Antibody Engineering," Curr. Op. Struct. Biol. 2(4):593-596.
Rai, K et al. (2000). "Chronic Lymphocytic Leukemia," Chap. 72 in Hematology, Basic Principles and Practice, Hoffman et al. eds., 3rd ed., Churchill Livingstone, Philadelphia, PA, pp. 1350-1362.
Ramaswamy, S. et al. (Dec. 18, 2001, e-pub. Dec. 11, 2001). "Multiclass cancer diagnosis using tumor gene expression signatures," Proc. Natl. Acad. Sci. U.S.A. 98(26):15149-15154.
Ravetch, J.V. et al. (1991). "FC Receptors," Annu. Rev. Immunol .9:457-492.
Reichmann, L. et al. (Mar. 24, 1988). "Reshaping Human Antibodies for Therapy," Nature 332(6162):323-329.
Saetre et al. Molecular Brain Research. 2004. 126: 198-206. *
Sambrooks, J. (1989). "Molecular Cloning: A Laboratory Manual", second edition. Cold Spring Harbor Laboratory, pp. v-xxxii. (Table of Contents Only.).
Schier, R. et al. (1996). "Identification of Functional and Structural Amino-Acids Residues by Parsimonious Mutagenesis," Gene 169(2):147-155.
Shi, X. et al. (Nov. 16, 2008). "Identification of a Diagnostic Gene Signature for SGN-40, Anti-CD40 Monoclonal Antibody, in Pre-Clinical NHL Models and the Role of FAS in SGN-40 Mediated Apoptosis," 50th Annual Meeting of the American Society of Hematology, San Francisco, California, Dec. 6 through 9, 2008, three pages. Blood 112(11):565.
Sidhu, S.S. et al. (Apr. 2004). "Phage-Displayed Antibody Libraries of Synthetic Heavy Chain Complementarity Determining Regions," J. Mol. Biol. 338(2):299-310.
Singh et al. Proceedings of the New Zealand Society of Animal Production. 2004. 64: 8-10. *
Singleton et al., (1994). Dictionary of Microbiology and Molecular Biology, 2nd ed., J. Wiley & Sons, Chichester, United Kingdom, pp. vii-xiii. (Preface and Notes to the User Only.).
Smiraglia et al. Human Molecular Genetics. 2001. 10: 1413-1419. *
Smith, M.B. et al. (1992). "Advanced Organic Chemistry Reactions, Mechanisms and Structure," 4th ed., John Wiley & Sons, New York, N.Y, pp. xiii, xiv. (Table of Contents Only.).
The Free Dictionary definition for "measuring", printed on Mar. 18, 2014, available via url: . *
The Free Dictionary definition for "measuring", printed on Mar. 18, 2014, available via url: < thefreedictionary.com/measuring>. *
The Free Dictionary, definition for "determining", printed on Mar. 4, 2014, available via url: . *
The Free Dictionary, definition for "determining", printed on Mar. 4, 2014, available via url: < thefreedictionary.com/determining>. *
Thomas, P.S. (Sep. 1980). "Hybridization fo Denatured RNA and Small DNA Fragments Transferred to Nitrocellulose," Proc. Natl. Acad. Sci. USA 77(9):5201-5205.
Turner, J.G. et al. (Jan. 1, 2001). "Anti-CD40 Antibody Induces Antitumor and Antimetasatic Effects: The Role of NK Cells," Journal of Immunology, American Association of Immunologist 166(1):89-94.
Tzankov et al. J Clin Pathol. 2003. 56: 747-752. *
Tzankov, A. et al. (2003). "Prognostic Signiicance of CD44 Expression in Diffuse Large B Cell Lymphoma of Activated and Germinal Centre B Cell-Like Types: A Tissue Microarray Analysis of 90 Cases," J. Clin. Pathol. 56:747-752.
Van Besien, K. et al. (2000). "Clinical Manifestations, Staging and Treatment of Non-Hodgkin's Lymphoma," Chapter 70 in Hematology, Basic Principles and Practice, 3rd ed. Hoffman et al. eds., Churchill Livingstone, Philadelphia, PA, pp. 1293-1338.
Written Opinion mailed on Aug. 24, 2010, for PCT Application No. PCT/US2010/031528, filed on Apr. 17, 2010, nine pages.
Written Opinion mailed on Mar. 11, 2009, for PCT Application No. PCT/US2008/082920, filed on Nov. 7, 2008, five pages.
Yelton, D.E. et al. (Aug. 15, 1995). "Affinity Maturation of the BR96 Anti-Carcinoma Antibody by Codon-Based Mutagenesis," J Immunol. 155(4):1994-2004.
Zhou, H. et al. (2005)"Regularization and Variable Selection Via the Elastic Net;" J. R. Statist. Soc. B. 67(2):301-320.

Also Published As

Publication number Publication date
WO2009062125A1 (en) 2009-05-14
CA2704499C (en) 2020-03-10
AU2008323701A1 (en) 2009-05-14
US20110104671A1 (en) 2011-05-05
CN101910414A (en) 2010-12-08
IL205391A0 (en) 2010-12-30
BRPI0820707A2 (en) 2015-06-16
JP2014057588A (en) 2014-04-03
EP2222875A1 (en) 2010-09-01
EP2222875B1 (en) 2014-08-20
JP2011504102A (en) 2011-02-03
JP5730576B2 (en) 2015-06-10
KR101620642B1 (en) 2016-05-12
IL205391A (en) 2015-08-31
KR20100095564A (en) 2010-08-31
AU2008323701B2 (en) 2015-03-26
CN101910414B (en) 2016-01-13
CA2704499A1 (en) 2009-05-14
MX2010005080A (en) 2010-07-28

Similar Documents

Publication Publication Date Title
US9068230B2 (en) Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies
US9617600B2 (en) Methods for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies
US9139878B2 (en) Methods for diagnosing and treating systemic lupus erythematosus
US20100279298A1 (en) Methods and Compositions For Detecting Autoimmune Disorders
AU2013202908A1 (en) Methods for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies
AU2013203418A1 (en) Methods and compositions for detecting autoimmune disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENTECH, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DORNAN, DAVID;BURINGTON, BART;REEL/FRAME:024971/0403

Effective date: 20100817

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8